ModernaTX, Inc. et al v. Pfizer Inc. et al

Filing 1

COMPLAINT against BioNTech US Inc., BioNTech SE, Pfizer Inc., BioNTech Manufacturing GmbH Filing fee: $ 402, receipt number AMADC-9469742 (Fee Status: Filing Fee paid), filed by Moderna US, Inc., ModernaTX, Inc.. (Attachments: #1 Exhibit 1, #2 Exhibit 2, #3 Exhibit 3, #4 Exhibit 4, #5 Exhibit 5, #6 Exhibit 6, #7 Exhibit 7, #8 Exhibit 8, #9 Exhibit 9, #10 Civil Cover Sheet, #11 Category Form)(Lee, William)

Download PDF
EXHIBIT 1 US010898574B2 ( 12 ) deUnited States Patent Fougerolles et al . ( 10 ) Patent No.: US 10,898,574 B2 (45 ) Date of Patent : * Jan . 26 , 2021 ( 54 ) DELIVERY AND FORMULATION OF ( 58 ) Field of Classification Search None See application file for complete search history . ENGINEERED NUCLEIC ACIDS ( 71 ) Applicant: Moderna TX , Inc. , Cambridge, MA References Cited U.S. PATENT DOCUMENTS ( 56) (US ) (72) Inventors : Antonin de Fougerolles , Waterloo ( BE ) ; Sayda M. Elbashir , Cambridge, MA ( US ) 5,034,506 A 5,426,180 A 5,489,677 5,512,439 5,591,722 5,637,459 5,639,873 5,641,400 5,789,578 5,808,039 5,989,911 6,022,715 6,022,737 6,248,268 6,303,378 6,423,492 ( 73 ) Assignee : Moderna TX , Inc. , Cambridge , MA ( US ) ( * ) Notice: Subject to any disclaimer, the term of this patent is extended or adjusted under 35 U.S.C. 154 (b ) by 0 days. This patent is subject to a terminal dis claimer . (21 ) Appl. No .: 15 /927,730 Mar. 21 , 2018 (22) Filed : Prior Publication Data ( 65 ) US 2019/0060458 A1 6,511,832 B1 6,521,411 B2 7,691,569 B2 8,075,780 B2 8,093,367 8,664,194 8,680,069 8,691,750 8,710,200 8,716,465 8,802,438 Feb. 28 , 2019 Related U.S. Application Data ( 60 ) Continuation of application No. 15 / 379,284 , filed on May 18 , 2013 , now abandoned, which is continuation of application No. 13 /437,034 , filed on Apr. 2 , 2012 , now Pat. No. 8,710,200 . A61K 47/18 A61K 31/7088 ( 2017.01) ( 2006.01 ) A61K 31/7115 ( 2006.01 ) A61K 38/19 A61K 38/48 ( 2006.01 ) ( 2006.01 ) ( 2006.01 ) ( 2006.01 ) ( 2006.01 ) (2006.01 ) ( 2006.01 ) ( 2006.01 ) A61K 47/22 A61K 4728 CO7K 14/535 C12N 15/67 C12N 15/87 C12N 9/64 A61K 48/00 ( 2006.01 ) ( 52) U.S. CI . CPC A61K 47/18 ( 2013.01 ) ; A61K 31/7088 ( 2013.01 ) ; A61K 31/7115 ( 2013.01 ) ; A6IK 38/193 (2013.01 ) ; A61K 38/4846 ( 2013.01 ) ; A61K 47/22 ( 2013.01 ) ; A61K 47/28 ( 2013.01 ) ; CO7K 14/535 (2013.01 ) ; C12N 9/644 (2013.01 ) ; C12N 15867 ( 2013.01 ) ; C12N 15/87 (2013.01 ) ; A61K 48/00 ( 2013.01 ) ; C12N 2310/335 (2013.01) 6/1997 Burke et al . 6/1997 Barascut et al . 6/1997 Kaltenbach et al. 8/1998 Burton et al . 9/1998 Reddy et al . 11/1999 2/2000 2/2000 6/2001 Fournier et al . Merenkova et al . Niven et al . Cook 10/2001 Bridenbaugh et al . 7/2002 Harbron 1/2003 Guarino et al . 2/2003 Hecker et al . 4/2010 Wohlgemuth et al . 12/2011 Pearce 1/2012 Kore et al. 3/2014 de Fougerolles et al. 3/2014 de Fougerolles et al. 4/2014 Constien et al . 4/2014 Schrum et al. 5/2014 Rossi et al . 8/2014 Rossi et al . 9/2014 Schrum et al. FOREIGN PATENT DOCUMENTS CA CA 2028849 A1 2473135 A1 9/1991 6/2003 (Continued ) OTHER PUBLICATIONS ( 60) Provisional application No. 61 / 470,451 , filed on Mar. 31 , 2011 . ( 1996 Sanghvi et al . 1/1997 Montgomery et al . 4/1996 Hornes et al . ( Continued ) division of application No. 14 /337,513 , filed on Jul. 22 , 2014 , now Pat. No. 9,533,047 , which is a continuation of application No. 13 / 897,362 , filed on a B2 B2 B2 B2 B2 B2 B2 8,822,663 B2 Dec. 14 , 2016 , now Pat . No. 9,950,068 , which is a (51 ) Int. Ci . A A A A A A A A A A A B1 B1 B1 7/1991 Summerton et al . 6/1995 Kool Lorenzi, et al . ( 2010 ) “ Intranasal vaccination with messenger RNA as a new approach in gene therapy: Use against tuberculosis ” , BMC Biotechnology, 10 : 77 ( 11 pages ). * Mignone, et al . ( 2002 ) " Untranslated regions of mRNAs" , Genome Biology, 3 ( 3 ) : reviews0004.1-0004.10 , 10 pages.* Sonoke , et al . ( 2008 ) “ Tumor Regression in Mice by Delivery of Bcl - 2 Small Interfering RNA with Pegylated Cationic Liposomes ”, Cancer Research , 68 ( 21 ) : 8843-51 . * Belanger, et al. ( 2010 ) “ Characterization of hMTr1, a Human Capl 2 ' - O -Ribose Methyltransferase ” Journal of Biological Chemistry , 285 (43 ) : 33037-44 . * (Continued ) Primary Examiner Robert M Kelly ( 74 ) Attorney, Agent, or Firm — Clark & Elbing LLP ( 57 ) ABSTRACT Provided are formulations, compositions and methods for delivering biological moieties such as modified nucleic acids into cells to modulate protein expression. Such compositions and methods include the delivery of biological moieties, and are useful for production of proteins. 10 Claims , 20 Drawing Sheets Specification includes a Sequence Listing . US 10,898,574 B2 Page 2 ( 56 ) References Cited U.S. PATENT DOCUMENTS 8,883,506 8,898,864 8,969,353 8,980,864 8,999,380 9,050,297 9,061,059 9,089,604 9,095,552 9,107,886 9,114,113 9,149,506 B2 B1 B2 B2 B2 B2 B2 B2 B2 B2 B2 B2 9,533,047 9,675,668 9,803,177 9,872,900 10,022,435 B2 B2 B2 B2 B2 9,428,535 B2 10,064,935 B2 10,286,086 B2 2001/0025097 2002/0001812 2002/0016450 2002/0062017 2002/0114784 2002/0130430 A1 A1 A1 Al A1 A1 2002/0153312 A1 2002/0164635 A1 2003/0120035 A1 2003/0170876 2003/0170891 2003/0180779 2004/0038278 2004/0142433 2004/0220127 2004/0259097 2005/0003496 2005/0130196 2006/0058266 2006/0121441 2006/0223081 2007/0020678 2007/0037148 2007/0037770 2007/0244062 2007/0281336 A1 Al A1 Al Al A1 Al A1 Al Al Al A1 A1 Al Al A1 A1 2008/0076910 A1 2008/0171711 A1 2008/0274463 A1 2008/0311140 A1 2009/0215125 A1 2009/0264511 A1 2009/0286852 A1 2010/0015232 2010/0047261 2010/0086922 2010/0159456 2010/0178272 2010/0183639 2010/0255574 2010/0261228 2010/0261231 2010/0317532 2011/0130440 2011/0143397 2011/0218170 2011/0244026 2011/0281938 2012/0046346 2012/0100136 2012/0129261 2012/0251618 2012/0309053 2013/0046083 2013/0046084 A1 A1 Al Al A1 A1 A1 A1 A1 A1 Al A1 A1 A1 Al Al Al Al A1 Al Al Al 11/2014 Rossi et al . 12/2014 Porter 3/2015 Mahon et al . 3/2015 4/2015 6/2015 6/2015 7/2015 8/2015 8/2015 8/2015 10/2015 8/2016 Hoge et al . Bancel et al. Chakraborty et al. Chakraborty et al . Chakraborty et al. Chakraborty et al. Chakraborty et al. Chakraborty et al. Chakraborty et al. de Fougerolles et al . 6/2017 10/2017 1/2018 7/2018 9/2018 Bancel et al. Rossi et al . Ciaramella et al . Ciaramella et al . Ciaramella et al . 1/2017 de Fougerolles et al . 2013/0052721 2013/0058894 2013/0115272 2013/0115274 2013/0123481 2013/0156849 2013/0165504 2013/0197068 2013/0203115 2013/0244282 2013/0245103 2013/0245105 2013/0245106 2013/0251618 2013/0259923 2013/0259924 2014/0010861 2014/0105964 2014/0105966 2014/0147454 2014/0200261 2014/0206752 A1 A1 A1 Al Al Al A1 A1 Al Al A1 A1 A1 Al Al Al A1 A1 Al A1 A1 Al 2014/0206852 A1 5/2019 Roy et al. 9/2001 Sheridan et al . 1/2002 Smith et al . 2014/0243399 2014/0273230 2014/0275227 2014/0343129 5/2002 Hecker et al . 2014/0371302 A1 2/2002 Laugharn et al. 8/2002 Li et al . 9/2002 Castor 10/2002 Gjerde et al . 11/2002 Salerno 6/2003 Gao et al . 9/2003 Widner et al . 9/2003 McSwiggen 9/2003 Lofton -Day et al. 2/2004 Tzertzinis et al . 7/2004 Padgett et al . 11/2004 Sternberg et al . 12/2004 De Backer et al . 1/2005 McGall et al . 6/2005 Hofstadler et al . 3/2006 Manoharan et al . 6/2006 Spira 10/2006 Jarrell et al . 1/2007 Ault - Riche et al . 2/2007 Fong et al. 2/2007 Gryaznov et al . 10/2007 Laux et al . 12/2007 Jendrisak et al . 3/2008 Takkellapati et al . 7/2008 Hoerr et al . 11/2008 Chen et al . 12/2008 Lee et al . 8/2009 Reed et al. 10/2009 11/2009 1/2010 2/2010 4/2010 de Fougerolles et al . Besenbacher et al . Hoerr et al . Bryant et al . 7/2010 10/2010 10/2010 10/2010 12/2010 6/2011 6/2011 Uhlmann et al . Rosen et al . Kariko et al. 6/2010 Albitar 7/2010 Hartmann et al . Gharib et al. Kore et al . Liu et al . Manoharan et al . Kariko et al . 2015/0005372 2015/0017211 2015/0030576 2015/0050354 2015/0050738 2015/0051268 2015/0056253 2015/0064235 2015/0064236 2015/0064725 2015/0086614 2015/0111248 2015/0140120 2015/0141269 2015/0141499 2015/0166616 2015/0307542 A1 2016/0017313 2016/0024140 2016/0024141 2016/0024492 2016/0024547 2016/0032273 2016/0038612 2016/0177295 2016/0194368 2016/0194625 2016/0237108 2016/0244742 2016/0326575 2016/0354490 2016/0354491 2016/0354492 2016/0354493 2016/0367702 2017/0088888 2017/0136131 2017/0136132 A1 5/2013 5/2013 5/2013 6/2013 de Fougerolles et al. Knopov et al . de Fougerolles et al. de Fougerolles et al. 6/2013 Bancel et al . 8/2013 Kariko et al . 8/2013 Schrum et al . 9/2013 Schrum et al . 9/2013 de Fougerolles et al. 9/2013 de Fougerolles et al . 9/2013 de Fougerolles et al. 9/2013 10/2013 10/2013 1/2014 4/2014 4/2014 5/2014 7/2014 7/2014 7/2014 Li et al . Bancel et al . Bancel et al . Bancel et al . Bancel et al . Bancel et al . Chakraborty et al . Hoge et al . Afeyan et al . Hoge et al . 8/2014 Schrum et al . 9/2014 Chen et al . 9/2014 Hoge et al . 11/2014 12/2014 1/2015 1/2015 1/2015 2/2015 2/2015 2/2015 2/2015 3/2015 3/2015 3/2015 3/2015 de Fougerolles et al. Afeyan et al . Hoge et al . de Fougerolles et al. Bancel Bouchon et al . Ozsolak et al . Bancel et al . Bancel et al . Bancel et al . Bancel et al . Schrum et al. Bancel et al . 4/2015 Bancel et al . 5/2015 McCauley et al. 5/2015 Soldatov et al . 5/2015 Bancel et al . 6/2015 Bancel et al . 6/2015 Roy et al. 7/2015 Wang et al . 10/2015 Roy et al. 1/2016 Spivak et al . 1/2016 Issa et al . 1/2016 Issa et al . 1/2016 Issa et al . 1/2016 Bancel et al . 2/2016 Shahrokh et al . 2/2016 7/2016 7/2016 8/2016 8/2016 Hoge et al . Hoge et al . Hoge et al . Fraley et al . Linnarsson et al . 6/2016 Rudolph et al. 11/2016 Von Der Mulbe et al . 12/2016 12/2016 12/2016 12/2016 12/2016 3/2017 5/2017 Roy et al. Roy et al . Roy et al. Roy et al. Hoge et al . El - Sagheer et al. Roy et al. 5/2017 Roy et al . 6/2017 Roy et al. FOREIGN PATENT DOCUMENTS 11/2011 Schaub et al . 2/2012 Rossi et al . 4/2012 Patel et al . 5/2012 Eberwine et al . 2/2013 Brown et al . 2/2013 Brown et al . Al A1 A1 A1 A1 A1 Al Al A1 Al A1 Al A1 A1 A1 A1 A1 A1 A1 A1 2017/0175129 Al 9/2011 Thottassery et al . 12/2012 Wellings A1 A1 A1 A1 A1 A1 Al Al A1 Al Al Al A1 A1 Al A1 2015/0167017 Al 2015/0211039 Al 10/2011 Guild et al . 10/2012 Schrum et al . A1 A1 A1 A1 2/2013 Hollander et al . 3/2013 Maldonado et al. ? 0366400 1083232 1611899 1619254 1383556 A2 B1 A1 A1 B9 1831160 B1 2092064 B1 5/1990 2/2005 1/2006 1/2006 3/2008 6/2010 9/2010 US 10,898,574 B2 Page 3 ( 56 ) References Cited FOREIGN PATENT DOCUMENTS EP EP 2377938 A1 2484770 A1 EP 2188379 B1 EP JP RU WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO 2548960 A1 2011-130725 A 2540017 C2 WO - 91 / 05058 Al WO - 93 / 03052 A1 WO - 93 / 13121 WO - 98/ 05673 WO - 99/ 14346 WO - 0042175 WO - 01 / 55306 Al A1 A2 A1 A2 WO - 02/44399 A2 WO - 03 /039523 A2 WO - 03 /051881 WO - 2004 /020575 WO - 2004 /020576 WO - 2004 /064782 WO - 2006 /015445 WO - 2007 /024708 Al A2 A2 A2 A1 A2 WO - 2007 /089607 WO - 2007 / 120863 WO - 2008 /039669 WO - 2008 /045505 A2 A2 Al A2 WO - 2008 /083949 A2 WO - 2008 / 120016 A1 WO - 2008 / 156829 A2 WO - 2009 /042971 WO - 2009 /051451 WO - 2009 / 127060 WO - 2009 / 127230 WO - 2009 / 147519 WO - 2009 / 149253 WO - 2010 /014895 WO - 2010 /017510 WO - 2010 /042877 WO - 2010 /054401 WO - 2010 / 109289 WO - 2010 / 144740 WO - 2011 /005850 WO - 2011 /012316 WO - 2011 /068810 WO - 2011 /071931 WO - 2011 / 127933 WO - 2011 / 130624 WO - 2011 / 133868 WO - 2011 / 140627 A2 A2 A1 A1 Al A2 A2 A1 A1 A1 A1 A1 A1 A2 A1 A2 A1 A2 A2 A1 WO - 2012 /019168 A2 WO - 2012 /077080 A1 WO - 2012 / 135805 A2 WO - 2012 / 138530 A1 WO - 2012 / 158736 A1 WO - 2012 / 164565 Al WO - 2013 /036748 WO - 2013 /039857 WO - 2013 /039861 WO - 2013 /052523 WO - 2013 /064911 WO - 2013 /090186 WO - 2013 /090294 WO - 2013 /090648 WO - 2013 /090897 WO - 2013 /096709 WO - 2013 / 101690 WO - 2013 / 103659 WO - 2013 / 113326 WO - 2013 / 113501 WO - 2013 / 113502 WO - 2013 / 130161 WO - 2013 / 151663 WO - 2013 / 151664 WO - 2013 / 151665 WO - 2013 / 151666 WO - 2013 / 151667 A1 A1 A2 A1 A2 A1 A1 A1 A1 A2 A1 Al Al Al A1 Al A1 Al A2 A2 A1 10/2011 8/2012 1/2013 1/2013 7/2011 1/2015 4/1991 2/1993 7/1993 2/1998 3/1999 7/2000 8/2001 6/2002 5/2003 6/2003 3/2004 3/2004 8/2004 2/2006 3/2007 8/2007 10/2007 4/2008 4/2008 7/2008 10/2008 12/2008 4/2009 4/2009 10/2009 10/2009 12/2009 12/2009 2/2010 2/2010 4/2010 5/2010 9/2010 12/2010 1/2011 2/2011 6/2011 6/2011 10/2011 10/2011 WO WO WO WO WO WO WO WO WO WO WO WO WO WO - 2013 / 151668 WO - 2013 / 151669 WO - 2013 / 151670 WO - 2013 / 151671 A2 A1 A2 Al WO - 2014 /028429 WO - 2014 /081507 WO - 2014 /093574 WO - 2014 /093622 A2 Al A1 A2 WO - 2013 / 151672 A2 WO - 2013 / 151736 A2 WO - 2013 / 184976 A2 WO WO - 2014 /093924 Al WO - 2014 / 113089 A2 WO - 2014 / 144039 Al WO WO WO WO - 2014 / 144711 Al WO - 2014 / 144767 A1 WO - 2014 / 152027 A1 WO WO - 2014 / 152030 WO - 2014 / 152031 WO - 2014 / 152211 WO - 2014 / 152513 WO - 2014 / 152540 WO - 2014 / 152659 WO - 2014 / 152673 WO - 2014 / 164253 WO - 2015 /006747 WO - 2015 /034925 WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO WO A1 A1 Al Al Al Al Al Al A2 A1 WO - 2015 /034928 A1 WO - 2015 /038892 WO - 2015 /048744 WO - 2015 /051169 WO - 2015 /051173 WO - 2015 /051214 WO - 2015 /058069 Al A2 A2 A2 A1 Al WO - 2015 /070413 Al WO - 2015 /085318 WO - 2015 /089511 WO - 2015 / 101414 WO - 2015 / 101416 WO - 2015 / 105926 WO - 2015 / 196118 WO - 2015 / 196128 WO - 2015 / 196130 WO - 2016 /010840 WO - 2016 /011222 WO - 2016 /011226 WO - 2016 /034620 WO - 2016 /036902 WO - 2016 / 118724 WO - 2016 / 118725 A2 A2 A2 Al A1 A1 A2 A2 A1 A2 A1 A1 A1 Al A1 10/2013 10/2013 10/2013 10/2013 10/2013 10/2013 12/2013 2/2014 5/2014 6/2014 6/2014 6/2014 7/2014 9/2014 9/2014 9/2014 9/2014 9/2014 9/2014 9/2014 9/2014 9/2014 9/2014 9/2014 10/2014 1/2015 3/2015 3/2015 3/2015 4/2015 4/2015 4/2015 4/2015 4/2015 5/2015 6/2015 6/2015 7/2015 7/2015 7/2015 12/2015 12/2015 12/2015 1/2016 1/2016 1/2016 3/2016 3/2016 7/2016 7/2016 10/2011 11/2011 OTHER PUBLICATIONS 10/2012 Pyhtila , et al . ( 2008 ) “ Signal sequence- and translational independent mRNA localization to the endoplasmic reticulum ” , 2/2012 6/2012 10/2012 11/2012 12/2012 3/2013 3/2013 3/2013 4/2013 5/2013 6/2013 6/2013 6/2013 6/2013 6/2013 7/2013 7/2013 8/2013 8/2013 8/2013 9/2013 10/2013 10/2013 10/2013 10/2013 10/2013 RNA, 14 ( 3 ) : 445-453 .* Brand et al . , “ Biosynthesis of a Hypermodified Nucleotide in Saccharomyces carlsbergensis 17S and HeLa - Cell 18S Ribosomal Ribonucleic Acid , ” Biochem J. 169 ( 1 ) :71-7 ( 1978 ) ( 9 pages ) . Kormann et al . , “ Expression of therapeutic proteins after delivery of chemically modified mRNA in mice," Nat Biotechnol . 29 ( 2 ) : 154-7 ( including supplement) ( 2011 ) ( 6 pages ) . RNA Modification Database Entry for 1 -methylpseudouridine < https:// mods.rna.albany.edu/mods/modifications/view/55 >, retrieved on Feb. 26 , 2019 ( 1 page ). Second Examination Report for Australian Patent Application No. 2017279733 , dated May 2 , 2019 ( 3 pages ) . Valadi et al., “ Exosome -mediated transfer ofmRNAs and microRNAs is a novel mechanism of genetic exchange between cells , ” Nat Cell Biol . 9 ( 6 ) : 654-9 ( 2007 ) ( 17 pages ). Anderson et al . , “ Incorporation ofpseudouridine into mRNA enhances translation by diminishing PKR activation ,” Nucleic Acids Res. 38 ( 17 ) : 5884-92 ( 2010 ) . Anderson et al . , “ Nucleoside modifications in RNA limit activation of 2-5 '-oligoadenylate synthetase and increase resistance to cleav age by RNase L , ” Nucleic Acids Res. 39 ( 21 ) : 9329-38 ( 2011 ) ( 10 pages). US 10,898,574 B2 Page 4 ( 56 ) References Cited OTHER PUBLICATIONS Andries et al . , “ N1-methylpseudouridine - incorporated mRNA out performs pseudouridine -incorporated mRNA by providing enhanced protein expression and reduced immunogenicity in mammalian cell lines and mice , " J Control Release . 217 : 337-44 ( 2015 ) . Applied Biosystems DNA Synthesizer model 380B operation manual, 2001 (327 pages). Aviv et al . , “ Purification of Biologically Active Globin Messenger RNA by Chromatography on Oligothymidylic acid -Cellulose,” Proc Nat Acad Sci USA 69 ( 6 ) : 1408-1412 ( 1972 ) . Communication pursuant to Rule 164 ( 2 ) (b ) and Article 94 ( 3 ) EPC for European Application No. 12722942.5 , dated Mar. 9 , 2018 ( 11 pages). Derrigo et al . , “ RNA -protein interactions in the control of stability and localization of messenger RNA (review ), ” Int J Mol Med . 5 ( 2 ) : 111-23 ( 2000 ) . Decision of Rejection for Japanese Patent Application No. 2016 148493 , dated Oct. 16 , 2018 ( 6 pages ) . Fath et al . , " Multiparameter RNA and codon optimization : a stan dardized tool to assess and enhance autologous mammalian gene expression , ” PLoS One 6 ( 3 ) : e17596 ( 2011 ) ( 14 pages ) . Gilham , “ The Synthesis of Polynucleotide -Celluloses and Their Use in the Fractionation of Polynucleotides,” J Am Chem Soc 86 ( 22):4982 4985 ( 1964) . Grosjean, Modification and editing of RNA : historical overview and important facts to remember. Fine - Tuning of RNA Functions by Modification and Editing. Grosjean H , 1-22 ( 2005 ) . Hansen et al ., “ Circular RNA and miR - 7 in Cancer,” Cancer Res. 73 ( 18 ) : 5609-12 (2013 ) . Hansen et al ., “ Natural RNA circles function as efficient microRNA sponges,” Nature . 495 ( 7441 ) : 384-8 ( 2013 ) ( 7 pages ) . Henke et al . , " microRNA - 122 stimulates translation of hepatitis C virus RNA , ” EMBO J. 27 ( 24 ) : 3300-10 ( 2008 ) . Hikishima et al . , “ Synthesis of 1,8 -naphthyridine C - nucleosides and their base -pairing properties in oligodeoxynucleotides : thermally stable naphthyridine: imidazopyridopyrimidine base -pairing motifs , ” Angew Chem Int Ed . 44 : 596-8 ( 2005 ) . Irier et al . , “ Translational regulation of GluR2 mRNAs in rat hippocampus by alternative 3 ' untranslated regions, ” available in PMC Aug. 17 , 2009 , published in final edited form as : J Neurochem . 109 ( 2 ) : 584-594 ( 2009 ) ( 18 pages ). Jani et al . , “ In vitro transcription and capping of Gaussia luciferase mRNA followed by HeLa cell transfection ,” J Vis Exp . 61 : e3702 ( 2012 ) ( 9 pages ) . Karikó et al . , “ Phosphate -enhanced transfection of cationic lipid complexed mRNA and plasmid DNA , ” Biochim Biophys Acta . 1369 ( 2 ) : 320-34 ( 1998 ) . Karikó et al ., “ mRNA is an endogenous ligand for Toll - like receptor 3 ,” J Biol Chem . 279 ( 13 ) : 12542-50 ( 2004 ) . Karikó et al . , “ In vivo protein expression from mRNA delivered into adult rat brain , ” J Neurosci Methods. 105 ( 1 ) : 77-86 ( 2001 ) . Karikó et al . , “ Increased erythropoiesis in mice injected with submicrogram quantities of pseudouridine - containing mRNA encod ing erythropoietin ,” Mol Ther. 20 ( 5 ) : 948-953 ( 2012 ) . Karikó et al . , “ Naturally occurring nucleoside modifications sup press the immunostimulatory activity of RNA: Implication for therapeutic RNA development , ” Curr Opin Drug Discov Devel. 10 ( 5 ) : 523-532 ( 2007 ) . Kariko et al . , “ Generating the optimal mRNA for therapy : HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein -encoding mRNA , ” Nucleic Acids Res . 39 (21 ) :e142, DOI : 10.1093/nar / gkr695 ( 2011) ( 10 pages ). Karikó et al . , “ Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capac ity and biological stability .” Mol Ther. 16 ( 11 ) : 1833-40 ( 2008 ) . Karikó et al ., “ Suppression of RNA recognition by Toll- like recep tors : the impact of nucleoside modification and the evolutionary origin of RNA ,” Immunity. 23 ( 2 ) : 165-75 ( 2005 ) . Kluiver et al., “ Rapid generation ofMicroRNA Sponges for MicroRNA Inhibition ,” PLoS One . 7 ( 1 ) : E29275 ( 2012 ) ( 8 pages ). Kore et al . , “ Synthesis and application of 2 ' - fluoro - substituted cap analogs.” Bioorg Med Chem Letters. 17 : 5295-9 (2007) . Kormann et al . , “ Expression of therapeutic proteins after delivery of chemically modified mRNA in mice , " Nat Biotechnol . 29 ( 2 ) : 154-7 ( 2011 ) ( 6 pages ) . Kuwahara et al . , “ Molecular evolution of functional nucleic acids with chemical modifications,” Molecules . 15 ( 8 ) : 5423-44 ( 2010 ) . Li et al . , “ Effects of chemically modified messenger RNA on protein expression , ” Bioconjug Chem . 27 ( 3 ) : 849-53 (2016 ). Melton et al . , “ Efficient in vitro synthesis of biologically active RNA and RNA hybridization probes from plasmids containing a bacteriophage SP6 promoter, ” Nucleic Acids Res. 12 ( 18 ) : 7035-56 ( 1984 ) . Memczak et al . , “ Circular RNAs are a large class of animal RNAs with regulatory potency , ” Nature . 495 ( 7441 ) : 333-8 ( 2013 ) ( 10 pages ) Meyer et al . , “ Combinatorial recombination of gene fragments to construct a library of chimeras,” Curr Protoc Protein Sci . Chapter 26 : Unit 26.2 (2006 ) ( 17 pages). Miyoshi- Akiyama et al . , " Complete genome sequence of Strepto coccus pyogenes M1 476 , isolated from a patient with streptococcal toxic shock syndrome," J Bacteriology. 194 ( 19 ) : 5466 ( 2012 ) . Moretti et al . , “ Mechanism of translational regulation by miR- 2 from sites in the 5 ' untranslated region or the open reading frame,” RNA . 16 ( 12 ) :2493-502 ( 2010 ). Motorin , “ RNA modification , ” eLS . John Wiley & Sons , DOI : 10 . 1002/ 9780470015902.a0000528.pub3 ( 2015 ) ( 18 pages ) . Nakazato et al . , “ Purification of messenger RNA and heterogeneous nuclear RNA containing poly ( A ) sequences , ” Methods Enzymol. 29 :431-443 ( 1974 ) . Nielsen et al . , “ An mRNA is capped by a 2 ' , 5 ' lariat catalyzed by a group I - like ribozyme,” Science . 309 ( 5740 ) : 1584-7 ( 2005 ) . Notification of Reason for Rejection for Japanese Application No. 2016-148493 , dated May 22 , 2018 ( 6 pages ) . Olesiak et al ., “ The synthesis of di- and oligo -nucleotides contain ing a phosphorodithioate internucleotide linkage with one of the sulfur atoms in a 5 '-bridging position , ” Org Biomol Chem . 7 ( 10 ) : 2162-9 ( 2009 ) . Pardi et al . , “ Expression kinetics of nucleoside -modified mRNA delivered in lipid nanoparticles to mice by various routes," available in PMC Nov. 10 , 2016 , published in final edited form as : J Control Release. 217 : 345-51 ( 2015 ) ( 18 pages ) . Qiu et al . , “ Creating a flexible multiple microRNA expression vector by linking precursor microRNAs,” Biochem Biophys Res Commun . 411 ( 2 ) : 276-80 ( 2011). Rodriguez et al . , “ Magnetic poly ( styrene/divinylbenzene / acrylic acid ) -based hybrid microspheres for bio -molecular recognition ," Micro Nano Lett . 6 ( 6 ) : 349-352 ( 2011 ) . Semple et al . , “ Rational design of cationic lipids for siRNA deliv ery,” Nat Biotechnol . 28 ( 2 ) : 172-6 ( 2010 ) ( 26 pages ). Stewart et al., “ Effect of azide position on the rate of azido glucose -cyclooctyne cycloaddition , ” Journal of Carbohydrate Chem istry. 33 ( 7-8 ) : 408-19 ( 2014 ) . Takita et al . , “ Precise sequential DNA ligation on a solid substrate : solid -based rapid sequential ligation of multiple DNA molecules, " DNA Res. 20 (6 ) :583-92 ( 2013 ) . Tavernier et al ., “ mRNA as gene therapeutic: how to control protein expression , ” J Control Release. 150 ( 3 ) : 238-47 ( 2011 ) . Virnekäs et al . , “ Trinucleotide phosphoramidites: ideal reagents for the synthesis of mixed oligonucleotides for random mutagenesis, " Nucleic Acids Res. 22 ( 25 ) : 5600-7 ( 1994 ) . Warren et al . , “ Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA , ” Cell Stem Cell . 7 ( 5 ) : 618-30 ( 2010 ) . Weiss et al., “ Prophylactic mRNA vaccination against allergy,” Curr Opin Allergy Clin Immunol . 10 ( 6 ) : 567-74 ( 2010 ) ( 8 pages ) . Wilusz et al . , “ Molecular Biology. A circuitous route to noncoding RNA,” Science . 340 ( 6131 ) : 440-1 ( 2013 ) . Yamamoto et al., " Current prospects for mRNA gene delivery , ” Eur J Pharm Biopharm . 71 ( 3 ) : 484-9 ( 2009 ) . “ Autoimmune shares collapse on Colloral data in rheumatoid arthritis,” Pharma MarketLetter, Marketletter Publications Ltd. ISSN : 0951-3175 ( 1999 ) . US 10,898,574 B2 Page 5 ( 56 ) References Cited OTHER PUBLICATIONS Anderson, Bart R. , Dissertation : “ Nucleoside Modifications Sup press RNA Activation of Cytoplasmic RNA Sensors," Doctor of Philosophy, Cell & Molecular Biology, University of Pennsylvania, 2010 ( 197 pages) . Bell et al . , “ In trans T cell tolerance diminishes autoantibody responses and exacerbates experimental allergic encephalomyeli tis," J Immunol. 180 ( 3 ) : 1508-16 ( 2008 ) . El - Sagheer et al . , “ Click nucleic acid ligation: applications in biology and nanotechnology, ” Acc Chem Res . 45 ( 8 ) : 1258-67 ( 2012 ) . Examination Report for Canadian Application No. 2,831,613 , dated Sep. 20 , 2019 (4 pages ) . Goodnow , “ Pathways for self -tolerance and the treatment of auto immune diseases, ” Lancet. 357 ( 9274 ) : 2115-21 ( 2001 ) . Haseltine et al . , “ Rous sarcoma virus genome is terminally redun dant: the 5 ' sequence , ” Proc Natl Acad Sci USA . 74 ( 3 ) : 989-93 ( 1977 ) . Jawalekar et al . , " Oligonucleotide tagging for copper - free click conjugation , ” Molecules . 18 ( 7 ) : 7346-63 ( 2013 ) . Kraus et al . , “ Oral tolerance and inflammatory bowel disease,” Curr Opin Gatroenterol. 21 ( 6 ) : 692-6 ( 2005 ) . Kurib shi- Ohta et al . , “ Ar lication of oligo (dT )30 - latex for rapid purification of poly ( A) + mRNA and for hybrid subtraction with the in situ reverse transcribed cDNA,” Biochim Biophys Acta. 1156 (2):204 12 ( 1993 ) . Lietard et al . , “ New strategies for cyclization and bicyclization of oligonucleotides by click chemistry assisted by microwaves,” J Org Chem . 73 ( 1 ) : 191-200 ( 2008 ) . Pozzilli et al ., “ No effect of oral insulin on residual beta - cell function in recent - onset type I diabetes (the IMDIAB VII)," Diabetologia. 43 ( 8 ) : 1000-4 ( 2000 ) . Santner et al ., “ Efficient access to 3 ' - terminal azide -modified RNA for inverse click - labeling patterns, ” Bioconjug Chem . 25 ( 1 ) : 188-95 ( 2014 ) . Skyler et al . , “ Effects of oral insulin in relatives of patients with type 1 diabetes : The Diabetes Prevention Trial — Type 1 , ” Diabetes Care 28 ( 5 ) : 1068-76 ( 2005 ) . Weiner et al., “ Oral tolerance , ” available in PMC May 1 , 2012 , published in final edited form as : Immunol Rev. 241 ( 1 ) : 241-59 ( 2011 ) ( 14 pages ) . Lukaysky et al . “ Large -scale Preparation and Purification of Polyacrylamide- Free RNA Oligonucleotides," RNA. 10 ( 5 ) : 889-93 ( 2004 ) ( 6 pages ) . Notification of Reasons for Rejection for Japanese Patent Applica tion No. 2016-148493 , dated Aug. 4 , 2020 (49 pages ) . Stocher et al. “ Removal of Template DNA From cRNA Preparations by Combined Oligo (dT ) Affinity Chromatography and DNase I Digestion ,” Biotechniques. 36 ( 3 ) : 480-2 ( 2004 ) . Wang et al . “ Improving the stability of aptamers by chemical Nagata et al., “ Molecular cloning and expression of cDNA for human granulocyte colony -stimulating factor,” Nature . 319 (6052 ) : 415-8 ( 1986 ) . Notification of Reasons for Rejection for Japanese Application No. modification ,” Curr Med Chem 18 ( 27 ) : 4126-38 ( 2011 ) . 2019-025010 , dated Mar. 3 , 2020 ( 12 pages ) . * cited by examiner U.S. Patent Jan. 26 , 2021 Sheet 1 of 20 US 10,898,574 B2 98N12-5 ( TETA5 - LAP ) HO OH H HON OH ?? DLin - K- DMA ( 2,2-Dilinoleyl -4 - dimethylaminomethyl- ( 1,3 ) -dioxolane) ge DLin - KC2 - DMA 88 DLin - MC3.DMA ocus you GoH21 OH min Cotar ???21 a OHC10H21 OH JOH C10H21 PRIOR ART U.S. Patent Sheet 2 of 20 Jan. 26 , 2021 US 10,898,574 B2 FIG . 2 Only single cutters are shown in the map BUSHI (3006 ) Asel ( 182 ) ASISI (385 ) ***** PN01:64818 haMSH optHs2 Apal (2072 ) txn terminator Rpn txi terminator Smal ( 1153 ) Hindill ( 1187) ECORV (1728) HincII ( 1710 ) Sphl ( 1614 ). Noti ( 1599 ) Sac? ( 1370) U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 3 of 20 HEK293, 24- well , 250 ng Modified RNA/ well TH untreaNPAted -001-1NPA-001-1aPNPA-002-1 NPA-003-1 NPA-005-1NPA-005-1aPNPA-006-1 FIG . 3B HepG2 , 24 - well , 250 ng Modified RNA /well 25,00 untreatNPAed -001-1NPA-001-1aPNPA-002-1NPA-002-1ap VIN-1-600deNPA-005.1NPA-005-1aPNPA-006-1NPA-006-12P U.S. Patent Jan. 26 , 2021 Sheet 4 of 20 US 10,898,574 B2 HEK293 , 24 - well , 250 ng Modified RNA /well untreated WPA-002-1 NPA-003-1ap NP4003-1 HepG2 , 24 - well , 250 ng Modified RNA/ well untreatNPAed -001-1NPA-001-1ap NPA-002-1apNPA-003-1 NPA-OOG-Iap NPA-005-1 NPA-005-1aNPA-006-1 U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 5 of 20 FIG . SA HEK293, NPA-005, 24-well , n =4 2 1 1. HEK293 , NPA - 003, 24-well , n =4 3 ***** 2 1.5 1 0 control 250ng U.S. Patent Jan. 26 , 2021 Sheet 6 of 20 US 10,898,574 B2 HEK293, NPA -003, 24 -well, n = 4 8 6 0 NPA -003-250ng NPA -003-100ng NPA - 003-30ng U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 7 of 20 HEK293 , 96 - well , 60 ng Modified RNA/ well untreated NBA-003 ?NPA01315 NPA-015 HEK293 , 62.5 ng/well 25000 MmChFerIy intreated NPA-015 NPA-006 NPA-013 NPA-008 NPA-003 NPA-007 NPA-010 NPA-012 NPA-009 NPA-005 NPA-001 NPA-014 NPA-002 U.S. Patent Jan. 26 , 2021 Sheet 8 of 20 US 10,898,574 B2 HEK293,62.5ng / well untreated NPA-007NPA-012 NPA-003-2NPA-018NPA-010NPA-015NPA-019 HepG2 , 62.5 ng / well TTTT untreated NPA-009 NPANPA -002-002-2 NPA-013 NPA-014 NPA-001 NPA-008 NPA-012 NPA-005 NPANPA -003-003-2 NPA-007 NPA-006 NPA-010 NPA-015 U.S. Patent Jan. 26 , 2021 Sheet 9 of 20 US 10,898,574 B2 HepG2 , 62.5ng / well 35000 untreatecdontrol ill NPA-007NPA-012NPA-017NPA-003-2NPA-018NPA-010NPA-015NPA-019 U.S. Patent Jan. 26 , 2021 Sheet 10 of 20 US 10,898,574 B2 Human EPO Protein in Mouse Serum ( I.M. Injection Route) 10 0 Pgimi Epo -Lipoplex Luc-Lipoplex Epo - Saline Epo-LipoplexLuc-LipoplexEpo-SalineLucSaline F.Buf er Treatment Groups U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 11 of 20 FIG . 7B Human EPO Protein in Mouse Serum ( S.C. Injection Route ) Luc -Lipoplex Epo - Saline ml / Pg Epo-LipoplexLuc-LipoplexEpo-SalineLuc-Saline FBuffer Treatment Groups U.S. Patent Jan. 26 , 2021 Sheet 12 of 20 US 10,898,574 B2 In vivo Biophotoic Imaging ( I.M. Injection- Left) 5ug Biolumnesc Leit, 2 hrs Left 8 hrs Left 24 hrs Photon ( sec / ) 5.0x105 Aqa luc Naked-LUCLipoplex Lipoplex-Cytokine FormulationBuffer Treatment U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 13 of 20 FIG . 8B In vivo Biophotoic Imaging ( I.M. Injection - Right) 50ug Biolumnesc 101 1x1006 Photon ( sec / ) luc Naked-LUCLipoplex Lipoplex-Cytokine Right 2 hrs Right, 8 hrs Right, 24 hrs FormulationBuffer Treatment U.S. Patent Jan. 26 , 2021 Sheet 14 of 20 US 10,898,574 B2 In vivo Biophotoic Imaging ( S.C.Injection Route) 50ug Biolumnesc 001 b)(esjuoj ud dhe Naked-LucLipoplexLipoplex -luc -G-CSF FormulationBuffer Treatment U.S. Patent Jan. 26 , 2021 Sheet 15 of 20 US 10,898,574 B2 In vivo Biophotoic Imaging (1.V. Injection Route) 50ug Spleen Blouminesc T ) sec / Photon ( -Naked-LucLipoplex-lucLipoplex-G-CSF FormulatioBnuf er Treatment U.S. Patent Jan. 26 , 2021 Sheet 16 of 20 Human EPO Protein ( IM Injection Route) US 10,898,574 B2 U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 17 of 20 Avers LUCBuf er unsp . Averg N1 CSF G .' . Avers CSF G TGreaotmupns Avers G-CSFinSHeurmuamn (1.M.,I.V.S.C.IRoute)njection 5 20 4 3 2 1 S.C. Avers N1 CSF G Averg 120.0 10 .0 80.0 7W/8d40.0 20.0 U.S. Patent Jan. 26 , 2021 Sheet 18 of 20 US 10,898,574 B2 Human G - CSF Protein in Mouse Serum ( 1.M. Injection Route) FBuffer Luc Saline Luc Lipoplex G2 -CSF -Gen2-Saline G--Lipoplex CSFG -Gent Saline -CSF--Gen -Lipoplex G-CSF-Gent Treatment Groups U.S. Patent Jan. 26 , 2021 Sheet 19 of 20 US 10,898,574 B2 FIG . 11B Human G - CSF Protein in Mouse Serum ( S.C. Injection Route ) mL / pg Anagrams FBuffer Luc saline Luc lipoplex -Gen2-saline GCSF-saline GenGCSF 2-lipoplex -GentGCSF-Gen1-GCSFlipoplex U.S. Patent Jan. 26 , 2021 US 10,898,574 B2 Sheet 20 of 20 Human EPO Protein in Mouse Serum ( IM Injection Route ) FBuffer +30Epo %.RNAIMAX +10%Epo RNAIMAX +50%RNAIMAX Epo#10%LucRNAIMAX +30%Luc RNA?MAX +50%LucRNAIMAX Treatment Groups US 10,898,574 B2 1 2 DELIVERY AND FORMULATION OF ENGINEERED NUCLEIC ACIDS modulation of intracellular translation and processing of nucleic acids encoding polypeptides and therefore optimiz ing protein expression from the delivered modalities . The present invention addresses this need by delivering This application is a continuation of U.S. patent application Ser. No. 15 /379,284 , filed Dec. 14 , 2016 , entitled 5 pharmaceutical compositions which can contain modified Delivery and Formulation of Engineered Nucleic Acids , nucleic acids such as modified mRNA (mmRNA ) and may which is a continuation of U.S. patent application Ser. No. further include formulations to avoid the problems in the art. 14 /337,513 , filed Jul . 22 , 2014 , entitled Delivery and For SUMMARY OF THE INVENTION mulation of Engineered Nucleic Acids , which is a continu ation of U.S. patent application Ser. No. 13 / 897,362 , filed 10 Described herein are compositions and methods for deliv May 18 , 2013 , entitled Modified Polynucleotides for the ery of biological moieties , such as modified nucleic acids , Production of Factor IX , which is a continuation of U.S. engineered messenger RNA and isolated polynucleotides patent application Ser. No. 13 /437,034 , filed Apr. 2 , 2012 , into in order to modulate protein expression . now issued U.S. Pat. No. 8,710,200 , entitled Delivery and 15 Ancells isolated polynucleotide may comprise a sequence such Formulation of Engineered Nucleic Acids which claims as , but not limited to , SEQ ID NO : 4 , 7 , 8 and 12. The priority to U.S. Provisional Patent Application No. 61/470 , polynucleotide further comprise a 5'Cap1 structure and 451 , filed Mar. 31 , 2011 , entitled Delivery and Formulation a polyA tail ofmay approximately 160 nucleotides in length . of Engineered Nucleic Acids the contents, the contents of Further, the isolated polynucleotide may be formulated in a each is incorporated by reference in its entirety . 20 pharmaceutical composition. A polypeptide of interest may be produced in a cell , tissue or bodily fluid in a subject in REFERENCE TO SEQUENCE LISTING need thereof by administering to the subject a pharmaceu The present application is being filed along with a tical composition comprising a polynucleotide. The poly Sequence Listing in electronic format. The Sequence Listing nucleotide may comprise a sequence selected from the group is provided as a file entitled MO03USSQLST.txt created on 25 consisting of SEQ ID NO : 4 , 7 , 8 and 12. The polynucleotide May 17 , 2013 which is 17,058 bytes in size . The information may further comprise a 5'Cap1 structure and a poly -A tail of in electronic format of the sequence listing is incorporated approximately 160 nucleotides in length . The pharmaceutical composition may be formulated herein by reference in its entirety . where the formulation may be selected from , but is not 30 limited to , saline or a lipid formulation . The pharmaceutical FIELD OF THE INVENTION composition may be administered by any route of adminis The invention relates to delivery methods. These methods tration such as , but not limited to , intravenous, intramuscu are specifically useful in therapeutic delivery of modified lar, subcutaneous, and local administration. The lipid for mulation may be selected from , but is not limited to , such as , nucleic acids such as modified mRNA (mmRNA ). 35 but not limited to , liposomes , lipoplexes , copolymers such BACKGROUND OF THE INVENTION as PLGA and lipid nanoparticles The pharmaceutical composition may be administered at There are multiple problems with prior methodologies of a total dose of about 0.1 mg /kg to about 40 mg /kg. The total delivering pharmaceutical compositions in order to achieve dose may be administered by multiple administrations. The effective protein expression both for therapeutics and bio- 40 administration and / or the multiple administration may occur processing applications. For example, introduced DNA can on a schedule such as , but not limited to , three time a day, integrate into host cell genomic DNA at some frequency, twice a day, once a day, every other day, every third day, resulting in alterations and /or damage to the host cell weekly, biweekly, every three weeks, every four weekly , and genomic DNA . Alternatively, the heterologous deoxyribo- monthly nucleic acid (DNA ) introduced into a cell can be inherited by 45 The modified polypeptide may include a polynucleotide daughter cells (whether or not the heterologous DNA has modification such as , but not limited to , a nucleoside modi integrated into the chromosome) or by offspring. fication . The nucleoside modification may include , but is not In addition, there are multiple steps which must occur limited to , pyridin - 4 -one ribonucleoside, 5 - aza -uridine , after delivery but before the encoded protein is made which 2 - thio - 5 -aza -uridine, 2 - thiouridine, 4 - thio -pseudouridine, can effect protein expression. Once inside the cell, DNA 50 2 -thio -pseudouridine, 5 -hydroxyuridine, 3 -methyluridine , must be transported into the nucleus where it is transcribed 5 - carboxymethyl -uridine, 1 -carboxymethyl-pseudouridine, into RNA . The RNA transcribed from DNA must then enter 5 -propynyl -uridine, 1 -propynyl -pseudouridine, 5-taurinom the cytoplasm where it is translated into protein . Not only do ethyluridine, 1 - taurinomethyl -pseudouridine, 5- taurinom the multiple processing steps from administered DNA to ethyl-2 -thio - uridine, 1 - taurinomethyl - 4 - thio - uridine, protein create lag times before the generation of the func- 55 5 -methyl-uridine, 1 -methyl-pseudouridine, 4 - thio -1 -methyl tional protein , each step represents an opportunity for error pseudouridine, 2 -thio -1 -methyl-pseudouridine, 1 -methyl- 1 and damage to the cell . Further, it is known to be difficult to deaza -pseudouridine, 2 -thio - 1-methyl- 1-deaza-pseudouri obtain DNA expression in cells as frequently DNA enters a dine, dihydrouridine, dihydropseudouridine, 2 -thio 2 - thio -dihydropseudouridine, cell but is not expressed or not expressed at reasonable rates dihydrouridine, or concentrations. This can be a particular problem when 60 2 -methoxyuridine, 2 -methoxy - 4 -thio -uridine, 4 -methoxy DNA is introduced into primary cells or modified cell lines . pseudouridine, 4 -methoxy - 2 -thio -pseudouridine, 5 -aza -cyti Assuming the proper management of the foregoing, effec- dine , pseudoisocytidine, 3 -methyl -cytidine, N4 -acetylcyti tive delivery and achievement of therapeutically relevant dine , 5 - formylcytidine, N4 -methylcytidine, levels of proteins for a time sufficient to product clinical 5 -hydroxymethylcytidine, 1 -methyl -pseudoisocytidine, pyr 65 rolo - cytidine, pyrrolo -pseudoisocytidine, 2 -thio -cytidine, outcomes remains a significant hurdle . Consequently, there is a need in the art for the delivery of 2 -thio -5 -methyl-cytidine, 4 -thio -pseudoisocytidine, 4 -thio biological modalities to address pitfalls surrounding the 1 -methyl -pseudoisocytidine, 4 -thio - 1 -methyl-1 -deaza -pseu US 10,898,574 B2 3 4 doisocytidine, 1 -methyl- 1 -deaza -pseudoisocytidine, zebularine , 5 - aza -zebularine, 5 -methyl-zebularine, 5 -aza- 2-thio- after purification ) that contain mCherry mmRNA . FIG . 4A shows the screening results in HEK293 cells and FIG . 4B zebularine, 2 - thio -zebularine, 2 -methoxy -cytidine, shows the screening results in HepG2 cells . FIGS . 5A , 5B , and 5C are histograms showing in vitro 2 -methoxy - 5 -methyl -cytidine, 4 -methoxy -pseudoisocyti dine, 4 -methoxy -1-methyl-pseudoisocytidine, 2-aminopu- 5 screening results for nanoparticle formulations of DLin KC2 -DMA and 98N12-15 before and after purification. FIG . rine , 2 , 6 -diaminopurine, 7 -deaza -adenine, 7 -deaza -8 -aza 5A shows the screening results of 98N15-2 in HEK293 cells , adenine, 7 -deaza - 2 -aminopurine, 7 -deaza - 8 - aza - 2 FIGS . 5B and 5C shows the screening results of aminopurine, 7 -deaza - 2,6 -diaminopurine, 7 - deaza - 8 - aza - 2 , and DLin -KC2 -DMA in HEK293 cells . 6 - diaminopurine, 1 -methyladenosine, N6 -methyladenosine, FIGS . 6A , 6B , 6C , and 6D are histograms showing in N6 - isopentenyladenosine, N6-(cis - hydroxyisopentenyl )ad- 10 vitro screening for nanoparticle formulations of enosine , 2 -methylthio -N6- (cis -hydroxyisopentenyl) adenos DLin - DMA , DLinresults K DMA , DLin -KC2 -DMA, 98N12-5 , ine , N6 - glycinylcarbamoyladenosine, N6 - threonylcarbam C12-200 and DLin -MC3 - DMA that contain mCherry oyladenosine, 2 -methylthio -N6 -threonylcarbamoylade mmRNA . FIG . 6A shows the mean fluorescence intensity of nosine , N6 , N6 -dimethyladenosine, 7 -methyladenine, mCherry in HEK293 cells containing 60 ng of modified 2 -methylthio - adenine, and 2 -methoxy -adenine, inosine, 15 mCherry mRNA per well . FIGS . 6B and 6C show the mean 1 -methyl- inosine, wyosine, wybutosine, 7 -deaza - guanosine, fluorescence intensity of mCherry in HEK293 cells which 7 -deaza - 8 -aza - guanosine, 6 - thio - guanosine, 6 - thio - 7 -deaza- contained nanoparticles formulations having a concentration guanosine, 6 - thio -7 -deaza - 8 -aza - guanosine, 7 -methyl- of 62.5 ng /well of modified mCherry mRNA . FIGS . 6D and guanosine, 6 -thio -7 -methyl -guanosine, 7 -methylinosine, 6E show the mean fluorescence intensity of mCherry in 6 -methoxy -guanosine, 1 -methylguanosine, N2 -methyl- 20 HepG2 cells which contained nanoparticle formulations guanosine, N2 ,N2 -dimethylguanosine, 8 -oxo - guanosine, 7 -methyl - 8 -oxo - guanosine, 1 -methyl-6 - thio - guanosine, having a concentration of 62.5 ng /well of modified mCherry mRNA . N2 -methyl - 6 -thio - guanosine, and N2,N2-dimethyl-6 -thioFIGS . 7A and 7B are histograms showing in vivo screen guanosine, and combinations thereof. ing results of human erythropoietin in serum after the An increase in the level of a polypeptide of interest can be 25 administration of modified human erythropoietin mmRNA observed in tissue such as , but not limited to , the liver, or luciferase mmRNA in mice . FIG . 7A shows the concen spleen , kidney, lung , heart, peri - renal adipose tissue , thymus tration in pg/ml of human erythropoietin after intramuscular and muscle and / or in a bodily fluid such as , but not limited administration . FIG . 7B shows the concentration in pg/ml of to , peripheral blood, serum , plasma , ascites, urine, cerebro- human erythropoietin after subcutaneous administration . spinal fluid (CSF ) , sputum , saliva , bone marrow , synovial 30 FIGS . 8A , 8B , 8C , and 8D are histograms of in vivo fluid , aqueous humor, amniotic fluid , cerumen , breast milk , broncheoalveolar lavage fluid , semen, prostatic fluid, cowper's fluid or pre - ejaculatory fluid , sweat , fecal matter, hair, tears , cyst fluid, pleural and peritoneal fluid , pericardial screening results from biophotoic imaging. FIG . 8A is a histogram of bioluminescence (photon / sec) from the intra muscular injection of 5 ug in the left hind leg . FIG . 8B is a histogram of bioluminescence from the intramuscular injec fluid , lymph, chyme, chyle , bile , interstitial fluid , menses , 35 tion of 50 ug in the right hind leg . FIG . 8C is a histogram pus , sebum , vomit , vaginal secretions, mucosal secretion, showing in vivo screening results from biophotoic imaging stool water, pancreatic juice , lavage fluids from sinus cavi- after a subcutaneous injection of 50 ug . FIG . 8D is a ties , bronchopulmonary aspirates , blastocyl cavity fluid, and histogram showing in vivo screening results from biopho umbilical cord blood . The increased level can be observed in toic imaging after a administration of 50 ug intravenously. the present invention . Shown are the structures for 98N12-5 ( TETA5 -LAP ), DLin - DMA, DLin- K - DMA ( 2,2 -Dilinoleyl4 -dimethylaminomethyl- [ 1,3 ) -dioxolane ), DLin -KC2 human G - CSF in serum after the administration of modified G -CSF subcutaneously. the tissue and /or bodily fluid of the subject within 2 , 8 and / or 40 FIG . 9 is a histogram showing in vivo screening results 24 hours after administration . Further, the increased level for modified human G - CSF mmRNA administered intra can be determined from the level of a modified polypeptide muscularly, subcutaneously or intravenously in mice . in an exosome . FIG . 10 is a histogram showing in vivo screening results The details of various embodiments of the invention are for modified G - CSF administered intramuscularly, subcuta set forth in the description below. Other features, objects , 45 neously or intravenously. and advantages of the invention will be apparent from the FIGS . 11A and 11B are histograms showing in vivo description and the drawings, and from the claims . screening results of modified human G - CSF mmRNA administered intramuscularly or subcutaneously in mice . BRIEF DESCRIPTION OF THE FIGURES FIG . 11A shows the concentration in pg /ml of human G - CSF 50 in serum after the administration of modified G - CSF intra FIG . 1 illustrates lipid structures in the prior art useful in muscularly. FIG . 11B shows the concentration in pg /ml of DMA , DLin -MC3 - DMA and C12-200 . FIG . 12 is a histogram showing in vivo screening results 55 of human erythropoietin in serum after the administration of modified human erythropoietin mmRNA or luciferase mmRNA administered intramuscularly in mice . Unless defined otherwise, all technical and scientific terms used herein have the same meaning as those com 60 monly understood to one of ordinary skill in the art to which this invention pertains. FIG . 2 is a representative plasmid useful in the IVT reactions taught herein . The plasmid contains Insert 64818 , designed by the instant inventors. FIGS . 3A and 3B are histograms showing in vitro screening results for nanoparticle formulations of DLin-KC2DMA and 98N12-15 (before and after purification ) that contain mCherry mmRNA . FIG . 3A shows the screening DETAILED DESCRIPTION results in HEK293 cells and FIG . 3B shows the screening results in HepG2 cells . FIGS . 4A and 4B are histograms showing in vitro screen- 65 Described herein are compositions and methods for the ing results for mean fluorescence intensity for nanoparticle delivery of modified mRNA molecules in order to modulate formulations of DLin -KC2 -DMA and 98N12-15 ( before and protein expression . US 10,898,574 B2 5 6 As described herein and as in copending, co - owned applications International Application PCT /US2011 / 046861 filed Aug. 5 , 2011 and PCT /US2011 / 054636 filed Oct. 3 , 2011 , the contents of which are incorporated by reference 500 , 600 , 700 , 800 , 900 , 1,000 , 1,100 , 1,200 , 1,300 , 1,400 , 1,500 , 1,600 , 1,700 , 1,800 , 1,900 , 2,000 , 2,500 , and 3,000 , 4,000 , 5,000 , 6,000 , 7,000 , 8,000 , 9,000 , 10,000 , 20,000 , 30,000 , 40,000 , 50,000 , 60,000 , 70,000 , 80,000 , 90,000 or herein in their entirety, these modified nucleic acid mol- 5 up to and including 100,000 nucleotides ). ecules are capable of reducing the innate immune activity of In some embodiments , the modified mRNA of the present a population of cells into which they are introduced, thus invention includes from about 30 to about 100,000 nucleo increasing the efficiency of protein production in that cell tides (e.g. , from 30 to 50 , from 30 to 100 , from 30 to 250 , population. from 30 to 500 , from 30 to 1,000 , from 30 to 1,500 , from 30 10 to 3,000 , from 30 to 5,000 , from 30 to 7,000 , from 30 to Modified mRNAs (mmRNAs) This invention provides nucleic acids , including RNAs, 10,000 , from 30 to 25,000 , from 30 to 50,000 , from 30 to specifically mRNAs, that encode at least one polypeptide 70,000 , from 100 to 250 , from 100 to 500 , from 100 to and contain one or more modified nucleosides ( termed 1,000 , from 100 to 1,500 , from 100 to 3,000 , from 100 to “ modified nucleic acids” or “ modified nucleic acid mol- 5,000 , from 100 to 7,000 , from 100 to 10,000 , from 100 to ecules " or " engineered nucleic acids” ), which have useful 15 25,000 , from 100 to 50,000 , from 100 to 70,000 , from 100 properties including the lack of a substantial induction of the to 100,000 , from 500 to 1,000 , from 500 to 1,500 , from 500 innate immune response of a cell into which the mRNA is to 2,000 , from 500 to 3,000 , from 500 to 5,000 , from 500 to introduced . Because these mmRNAs enhance the efficiency 7,000 , from 500 to 10,000 , from 500 to 25,000 , from 500 to of protein production, intracellular retention of nucleic 50,000 , from 500 to 70,000 , from 500 to 100,000 , from acids , and viability of contacted cells , as well as possess 20 1,000 to 1,500 , from 1,000 to 2,000 , from 1,000 to 3,000 , reduced immunogenicity, these nucleic acids having these from 1,000 to 5,000 , from 1,000 to 7,000 , from 1,000 to properties are termed " enhanced ” nucleic acids or modified 10,000 , from 1,000 to 25,000 , from 1,000 to 50,000 , from RNAs herein . 1,000 to 70,000 , from 1,000 to 100,000 , from 1,500 to 3,000 , The term “ nucleic acid ,” in its broadest sense , includes from 1,500 to 5,000 , from 1,500 to 7,000 , from 1,500 to any compound and / or substance that comprise a polymer of 25 10,000 , from 1,500 to 25,000 , from 1,500 to 50,000 , from nucleotides linked via a phosphodiester bond . These poly- 1,500 to 70,000 , from 1,500 to 100,000 , from 2,000 to 3,000 , mers are often referred to as oligonucleotides. Exemplary nucleic acids include ribonucleic acids from 2,000 to 5,000 , from 2,000 to 7,000 , from 2,000 to 10,000 , from 2,000 to 25,000 , from 2,000 to 50,000 , from (RNAs), deoxyribonucleic acids ( DNAs ), threose nucleic acids ( TNAs), glycol nucleic acids (GNAs ), peptide nucleic 30 acids ( PNAs), locked nucleic acids ( LNAs) or hybrids thereof. They may also include RNAi - inducing agents , RNAi agents, siRNAs, shRNAs, miRNAs, antisense RNAs, ribozymes , catalytic DNA , RNA, RNAs that induce triple helix formation , aptamers, vectors , etc. 35 In preferred embodiments, the nucleic acid is one or more modified messenger RNAs (mmRNAs ). As described herein , in some embodiments the mmRNAs of the invention do not substantially induce an innate immune response of a cell into which the mRNA is introduced . 40 The mmRNA of the present invention may encode one or more polypeptides . Generally the polypeptides of interest are those which are naturally occurring in the mammalian genome. According to the present invention, the shortest length of 45 a modified mRNA , herein “ mmRNA , ” of the present disclosure can be the length of an mRNA sequence that may be sufficient to encode for a dipeptide. In another embodiment, the length of the mRNA sequence may be sufficient to encode for a tripeptide. In another embodiment, the length of 50 an mRNA sequence may be sufficient to encode for a tetrapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a pentapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for a hexapeptide. In 55 another embodiment, the length of an mRNA sequence may be sufficient to encode for a heptapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to encode for an octapeptide. In another embodiment, the length of an mRNA sequence may be sufficient to 60 encode for a nonapeptide. In another embodiment, the length 2,000 to 70,000 , and from 2,000 to 100,000) . Polypeptide Variants The mmRNA of the present invention may encode variant polypeptides, which have a certain identity with a reference polypeptide sequence, for example a wild type mRNA . The term “ identity ” as known in the art, refers to a relationship between the sequences of two or more peptides , as deter mined by comparing the sequences . In the art, “ identity ” also means the degree of sequence relatedness between peptides, as determined by the number of matches between strings of two or more amino acid residues. “ Identity " measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any ) addressed by a particular mathematical model or com puter program ( i.e. , “ algorithms” ). Identity of related pep tides can be readily calculated by known methods. Such methods include , but are not limited to , those described in Computational Molecular Biology , Lesk , A. M. , ed . , Oxford University Press, New York , 1988 ; Biocomputing: Informat ics and Genome Projects, Smith, D. W., ed . , Academic Press , New York , 1993 ; Computer Analysis of Sequence Data , Part 1 , Griffin , A. M. , and Griffin , H. G. , eds . , Humana Press, New Jersey, 1994 ; Sequence Analysis in Molecular Biology, von Heinje , G. , Academic Press , 1987 ; Sequence Analysis Primer, Gribskov , M. and Devereux, J. , eds . , M. Stockton Press, New York , 1991 ; and Carillo et al . , SIAM J. Applied Math . 48 , 1073 ( 1988 ) . In some embodiments, the polypeptide variant has the same or a similar activity as the reference polypeptide. Alternatively, the variant has an altered activity ( e.g. , increased or decreased ) relative to a reference polypeptide. Generally, variants of a particular polynucleotide or poly peptide of the invention will have at least about 40 % , 45 % , decapeptide. 92 % , 93 % , 94 % , 95 % , 96 % , 97 % , 98 % , 99 % or more of an mRNA sequence may be sufficient to encode for a Generally, the length of a modified mRNA of the present at least or greater than about 35 , 40 , 45 , 50 , 55 , 60 , 70 , 80 , 50% , 55 % , 60 % , 65 % , 70 % , 75 % , 80 % , 85 % , 90% , 91 % , sequence identity to that particular reference polynucleotide grams and parameters described herein and known to those invention is greater than about 30 nucleotides in length ( e.g. , 65 or polypeptide as determined by sequence alignment pro 90 , 100 , 120 , 140 , 160 , 180 , 200 , 250 , 300 , 350 , 400 , 450 , skilled in the art. US 10,898,574 B2 7 As recognized by those skilled in the art, protein frag- ments, functional protein domains, and homologous proteins are also considered to be within the scope of this invention . 8 approach, Oxford [ Oxfordshire ), Washington , D.C .: IRL Press, 1984 ; and Herdewijn , P. ( ed .) Oligonucleotide syn thesis : methods and applications, Methods in Molecular For example, provided herein is any protein fragment of a Biology, v. 288 ( Clifton , N.J. ) Totowa, N.J .: Humana Press , reference protein (meaning a polypeptide sequence at least 5 2005 ; both of which are incorporated herein by reference ). one amino acid residue shorter than a reference polypeptide sequence but otherwise identical) 10 , 20 , 30 , 40 , 50 , 60 , 70 , 80 , 90 , 100 or greater than 100 amino acids in length In The modified nucleosides and nucleotides used in the synthesis of modified RNAs disclosed herein can be pre pared from readily available starting materials using the which are about 40 % , about 50 % , about 60 % , about 70 % , about 80 % , about 90 % , about 95 % , or about 100 % identical reaction temperatures, times , mole ratios of reactants , sol vents , pressures , etc.) are given ; other process conditions can also be used unless otherwise stated . Optimum reaction conditions may vary with the particular reactants or solvent The modified nucleic acids and mmRNA may encode a et al . Nucleic Acids Research 22 ( 1 ) : 72-78 , 1994 ; Fukuhara way to permit the expression of the complex when introduced into the cell . As used herein , “ charged protein ” refers to a protein that carries a positive , negative or overall neutral the entire length of the molecule . Different nucleotide modi fications and / or backbone structures may exist at various positions in the nucleic acid . One of ordinary skill in the art another example, any protein that includes a stretch of about following general methods and procedures . It is understood 20 , about 30 , about 40 , about 50 , or about 100 amino acids 10 that where typical or preferred process conditions (i.e. , to any of the sequences described herein can be utilized in accordance with the invention . In certain embodiments, a protein sequence to be utilized in accordance with the 15 used, but such conditions can be determined by one skilled invention includes 2 , 3 , 4 , 5 , 6 , 7 , 8 , 9 , 10 , or more mutations in the art by routine optimization procedures. as shown in any of the sequences provided or referenced The manufacturing process herein can be monitored herein . according to any suitable method known in the art. For Targeting Moieties example , product formation can be monitored by spectro In embodiments of the invention , mmRNAs are provided 20 scopic means, such as nuclear magnetic resonance spectros to express a protein -binding partner or a receptor on the copy (e.g. , ' H or 13C ) infrared spectroscopy, spectropho surface of the cell , which functions to target the cell to a tometry (e.g. , UV - visible ), or mass spectrometry , or by specific tissue space or to interact with a specific moiety, chromatography such as high performance liquid chroma either in vivo or in vitro . Suitable protein -binding partners tography ( HPLC ) or thin layer chromatography. include antibodies and functional fragments thereof, scaffold 25 Modification of mRNAs proteins, or peptides . Provided are mmRNAs containing a translatable region Cell Penetrating Peptides and one , two , or more than two different modifications. The mmRNAs disclosed herein may encode a cell -penIn some embodiments, the chemical modifications can be etrating polypeptide. As used herein , “ cell- penetrating poly- located on the nucleobase of the nucleotide . peptide ” refers to a polypeptide which may facilitate the 30 In some embodiments, the chemical modifications can be cellular uptake of molecules . It is known in the art that located on the sugar moiety of the nucleotide . “ CPP ” refers to cell-penetration polypeptides and cell-penIn some embodiments, the chemical modifications can be etrating peptides . When used herein , it will be clarified as to located on the phosphate backbone of the nucleotide . which of either cell-penetrating polypeptides or cell -penPreparation of modified nucleosides and nucleotides used etrating peptides the abbreviation CPP refers to . 35 in the manufacture or synthesis of modified RNAs of the A cell-penetrating polypeptide of the present invention present invention can involve the protection and deprotec may contain one or more detectable labels . The polypeptides tion of various chemical groups. The need for protection and may be partially labeled or completely labeled throughout. deprotection , and the selection of appropriate protecting The mmRNA may encode the detectable label completely, groups can be readily determined by one skilled in the art. partially or not at all . The cell-penetrating peptide may also 40 The chemistry of protecting groups can be found, for include a signal sequence . As used herein , a “ signal example, in Greene, et al . , Protective Groups in Organic sequence” refers to a sequence of amino acid residues bound Synthesis, 2d . Ed . , Wiley & Sons , 1991 , which is incorpo at the amino terminus of a nascent protein during protein rated herein by reference in its entirety. translation . The signal sequence may be used to signal the Modified nucleosides and nucleotides can be prepared secretion of the cell -penetrating polypeptide. 45 according to the synthetic methods described in Ogata et al . Fusion Proteins Journal of Organic Chemistry 74 : 2585-2588 , 2009 ; Purmal fusion protein . The fusion protein may be created by oper- et al . Biochemistry 1 ( 4 ) : 563-568 , 1962 ; and Xu et al . ably linking a charged protein to a therapeutic protein . As Tetrahedron 48 ( 9 ) : 1729-1740 , 1992 , each of which are used herein , “ operably linked ” refers to the therapeutic 50 incorporated by reference in their entirety . protein and the charged protein being connected in such a Modified mRNAs need not be uniformly modified along electrical charge. Preferably , the therapeutic protein may be 55 will appreciate that the nucleotide analogs or other modifi covalently linked to the charged protein in the formation of cation (s) may be located at any position( s ) of a nucleic acid the fusion protein . The ratio of surface charge to total or such that the function of the nucleic acid is not substantially surface amino acids may be approximately 0.1 , 0.2 , 0.3 , 0.4 , decreased . A modification may also be a 5 ' or 3 ' terminal modification . The nucleic acids may contain at a minimum Synthesis of Modified mRNAs 60 one and at maximum 100 % modified nucleotides , or any Nucleic acids for use in accordance with the invention intervening percentage , such as at least 50 % modified may be prepared according to any available technique nucleotides , at least 80 % modified nucleotides, or at least including , but not limited to chemical synthesis, enzymatic 90% modified nucleotides . synthesis, which is generally termed in vitro transcription , For example, the mmRNAs may contain a modified enzymatic or chemical cleavage of a longer precursor, etc. 65 pyrimidine such as uracil or cytosine . In some embodiments, Methods of synthesizing RNAs are known in the art ( see , at least 5 % , at least 10 % , at least 25 % , at least 50% , at least e.g. , Gait , M. J. (ed . ) Oligonucleotide synthesis: a practical 80% , at least 90% or 100 % of the uracil in the nucleic acid 0.5 , 0.6 , 0.7 , 0.8 or 0.9 . US 10,898,574 B2 10 9 may be replaced with a modified uracil. The modified uracil can be replaced by a compound having a single unique structure , or can be replaced by a plurality of compounds tidine , 5 '- 0-( 1 - Thiophosphate ) -Guanosine, 5-0-( 1 - Thio phosphate ) -Uridine or 5 '-0-( 1 - Thiophosphate )-Pseudo uridine . Further examples of modified nucleotides and modified having different structures ( e.g. , 2 , 3 , 4 or more unique structures ). In some embodiments, at least 5 % , at least 10 % , 5 at least 25 % , at least 50 % , at least 80 % , at least 90 % or nucleotide combinations are provided below in Table 1 . 100% of the cytosine in the nucleic acid may be replaced TABLE 1 with a modified cytosine . The modified cytosine can be Modified Nucleotides Modified Nucleotide Combinations replaced by a compound having a single unique structure , or 10 can be replaced by a plurality of compounds having different 6 -aza -cytidine a -thio -cytidine/ 5 -iodo -uridine 2 - thio -cytidine a - thio -cytidine /N1-methyl -pseudo -uridine structures (e.g. , 2 , 3 , 4 or more unique structures ). a - thio -cytidine a - thio -cytidine / a -thio - uridine In some embodiments, modified nucleosides include pyri Pseudo - iso - cytidine a - thio -cytidine/ 5 -methyl -uridine din - 4 - one ribonucleoside, 5 -aza -uridine, 2 -thio - 5 - aza-uri 5 - aminoallyl -uridine a -thio -cytidine/pseudo -uridine Pseudo -iso -cytidine / 5 -iodo - uridine dine , 2 - thiouridine, 4 - thio -pseudouridine, 2- thio -pseudouri- 15 5 - iodo - uridine dine, 5 -hydroxyuridine , 3 -methyluridine, 5 -carboxymethyl- N1 -methyl-pseudouridine Pseudo - iso -cytidine /N1-methyl-pseudo uridine, 5,6 -dihydrouridine 1 -carboxymethyl-pseudouridine, 5 -propynyl uridine, 1 -propynyl -pseudouridine, 5 -taurinomethyluridine, 1 -taurinomethyl -pseudouridine, 5 - taurinomethyl- 2- thio -uri- a -thio -uridine 4 -thio -uridine dine , 1 - taurinomethyl - 4 - thio -uridine, 5 -methyl-uridine, 20 5 - hydroxy -uridine Deoxy -thymidine 1 -methyl-pseudouridine, 4 - thio - 1 -methyl -pseudouridine, 6 -aza -uridine 2 - thio - 1 -methyl-pseudouridine, 1 -methyl- 1-deaza -pseudou- Pseudo -uridine ridine, 2 - thio - 1 -methyl - 1 -deaza - pseudouridine, dihydrouri Inosine thio - guanosine dine, dihydropseudouridine, 2 -thio -dihydrouridine, 2 -thio a8 --oxo - guanosine dihydropseudouridine, 2 -methoxyuridine, 2 -methoxy -4- 25 06 -methyl-guanosine 7 -deaza - guanosine thio - uridine, 4 -methoxy -pseudouridine, and 4 -methoxy -2 No modification thio -pseudouridine. In some embodiments, modified N1 -methyl - adenosine nucleosides include 5 - aza -cytidine, pseudoisocytidine, 3 -methyl-cytidine, N4 - acetylcytidine, 5 - formylcytidine, N4 -methylcytidine, 5 -hydroxymethylcytidine, 1 -methyl 30 pseudoisocytidine, pyrrolo -cytidine, pyrrolo -pseudoisocyti dine, 2 - thio - cytidine , 2 - thio - 5 -methyl -cytidine, 4 -thio -pseu doisocytidine, 4 - thio - 1 -methyl -pseudoisocytidine, 4 - thio - 1 methyl - 1 - deaza -pseudoisocytidine, 1 -methyl- 1 -deaza 35 pseudoisocytidine, zebularine , 5 - aza - zebularine , 5 -methyl zebularine, 5 - aza - 2 - thio - zebularine , 2 - amino - 6 - Chloro -purine uridine Pseudo - iso -cytidine / a -thio -uridine Pseudo - iso -cytidine / 5 -methyl -uridine Pseudo - iso -cytidine /Pseudo -uridine Pyrrolo - cytidine Pyrrolo - cytidine /5 - iodo -uridine Pyrrolo -cytidine/N1-methyl-pseudo -uridine Pyrrolo -cytidine / a - thio - uridine Pyrrolo -cytidine / 5 -methyl -uridine Pyrrolo -cytidine /Pseudo -uridine 5 -methyl- cytidine / 5 - iodo -uridine 5 -methyl-cytidine /N1-methyl-pseudo - uridine 5 -methyl-cytidine / a - thio -uridine 5 -methyl -cytidine / 5 -methyl -uridine 5 -methyl -cytidine /Pseudo -uridine 5 -methyl-cytidine N6 -methyl- 2 -amino -purine 25 % Pseudo - iso -cytidine 6 - Chloro -purine 25 % N1-methyl-pseudo-uridine N6 -methyl -adenosine 25 % N1-Methyl-pseudo -uridine /75 % pseudo -uridine 5 -methyl-uridine a -thio - adenosine 8 - azido - adenosine 7 - deaza- adenosine 5 - iodo- cytidine 2 - thio -zebularine, In some embodiments, at least 25 % of the cytosines are yisopentenyl ) adenosine, 2-methylthio -N6-(cis-hydroxyisoN6 - threonylcarbamoyladenosine, 2 -methylthio -N6 -threonyl carbamoyladenosine, N6 , N - dimethyladenosine, 7 -methyl- 45 % , at least about 50 % , at least about 55 % , at least about 60% , at least about 65 % , at least about 70 % , at least about 75 % , at least about 80 % , at least about 85 % , at least about 50 90% , at least about 95 % , or about 100 % ) . In some embodiments , at least 25 % of the cytosines and 25 % of the uracils are replaced by a compound of Formula I - a ( e.g. , at least about 30 % , at least about 35 % , at least 55 about 40 % , at least about 45 % , at least about 50 % , at least about 55 % , at least about 60% , at least about 65 % , at least about 70 % , at least about 75 % , at least about 80 % , at least about 85 % , at least about 90 % , at least about 95 % , or about 100 % ). 60 Other components of nucleic acid are optional , and are 2 -methoxy -cytidine, 2 -methoxy - 5 -methyl -cytidine, replaced by a compound of Formula I - a ( e.g. , at least about 4 -methoxy -pseudoisocytidine, and 4 -methoxy - 1-methyl- 30% , at least about 35 % , at least about 40 % , at least about pseudoisocytidine. 40 45 % , at least about 50 % , at least about 55 % , at least about In other embodiments, modified nucleosides include 60% , at least about 65 % , at least about 70 % , at least about 2 -aminopurine , 2 , 6 -diaminopurine, 7 -deaza - adenine , 75 % , at least about 80 % , at least about 85 % , at least about 7 -deaza - 8 -aza -adenine, 7 -deaza - 2 -aminopurine, 7 - deaza -8- 90% , at least about 95 % , or about 100 % ) . aza - 2 - aminopurine, 7 -deaza -2,6 -diaminopurine, 7 -deaza - 8In some embodiments, at least 25 % of the uracils are aza - 2,6 -diaminopurine, l -methyladenosine, N6 -methylad- 45 replaced by a compound of Formula 1 - a (e.g. , at least about enosine, N6 - isopentenyladenosine, N6- (cis-hydrox 30% , at least about 35 % , at least about 40 % , at least about pentenyl) adenosine, N6 - glycinylcarbamoyladenosine, adenine , 2 -methylthio -adenine, and 2 -methoxy -adenine. In other embodiments, modified nucleosides include inos ine , 1 -methyl - inosine, wyosine , wybutosine, 7-deaza guanosine, 7 - deaza - 8 - aza -guanosine, 6 - thio -guanosine, 6 - thio - 7 - deaza -guanosine, 6 - thio - 7 -deaza - 8 -aza - guanosine , 7 -methyl- guanosine, 6 -thio - 7 -methyl -guanosine, 7 -methylinosine , 6 -methoxy - guanosine, 1 -methylguanosine, N2 -methylguanosine, N2 ,N2 - dimethylguanosine, 8 -oxo- guanosine, 7 -methyl - 8 -oxo - guanosine, 1 -methyl- 6 -thioethyl -6 - thio - guanosine. guanosine, N2-methyl -6 - thio -guanosine, and N2 N2-dim beneficial in some embodiments. For example, a 5 ' untrans lated region (UTR ) and /or a 3'UTR are provided, wherein In some embodiments, the nucleotide can be modified on either or both may independently contain one or more the major groove face and can include replacing hydrogen different nucleoside modifications . In such embodiments , on C - 5 of uracil with a methyl group or a halo group . 65 nucleoside modifications may also be present in the trans In specific embodiments , a modified nucleoside is 5'- 0- latable region. Also provided are nucleic acids containing a ( 1 - Thiophosphate)-Adenosine, 5'-0-( 1- Thiophosphate )-Cy Kozak sequence . US 10,898,574 B2 11 12 Linkers and Payloads The nucleobase of the nucleotide, which may be incorporated into a mmRNA , can be covalently linked at any chemically appropriate position to a payload, e.g. detectable agent or therapeutic agent. For example , the nucleobase can be deaza - adenosine or deaza -guanosine and the linker can be attached at the C - 7 or C - 8 positions of the deaza - adenosine or deaza -guanosine. In other embodiments , the nucleobase can be cytosine or uracil and the linker can be attached to the Labels , other than those described herein , are contem plated by the present disclosure, including , but not limited to , other optically -detectable labels. Labels can be attached to the modified nucleotide of the present disclosure at any position using standard chemistries such that the label can be removed from the incorporated base upon cleavage of the 5 cleavable linker. Terminal Architecture Modifications: 5 ' - Capping 10 N - 3 or C - 5 positions of cytosine or uracil. The term “ linker” as used herein refers to a group of atoms , e.g. , 10-1,000 atoms , and can be comprised of the atoms or groups such as , but not limited to , carbon , amino , alkylamino , oxygen , sulfur, sulfoxide, sulfonyl, carbonyl, 15 Linker and imine . The linker can be attached to a modified nucleo side or nucleotide on the nucleobase or sugar moiety at a first end , and to a payload , e.g. , detectable or therapeutic agent, at a second end . The linker may be of sufficient length as to not interfere with incorporation into a nucleic acid sequence . 20 Examples of chemical groups that can be incorporated into the linker include, but are not limited to , an alkyl, an alkene , an alkyne, an amido , an ether, a thioether or an ester group . The linker chain can also comprise part of a saturated , unsaturated or aromatic ring, including polycyclic and het- 25 eroaromatic rings wherein the heteroaromatic ring may be an aryl group containing one to four heteroatoms, N , O or S. Specific examples of linkers include , but are not limited to , unsaturated alkanes, polyethylene glycols , and dextran polymers . 30 For example , the linker can include, but is not limited to , ethylene or propylene glycol monomeric units, e.g. , diethylene glycol , dipropylene glycol, triethylene glycol , tripro- pylene glycol , tetraethylene glycol , or tetraethylene glycol . In some embodiments, the linker can include , but is not 35 limited to , a divalent alkyl, alkenyl, and / or alkynyl moiety . The linker can include an ester, amide , or ether moiety. Other examples include , but are not limited to , cleavable moieties within the linker, such as , for example, a disulfide bond ( S S ) or an azo bond ( N = N— ), which can be cleaved using a reducing agent or photolysis . When a cleavable bond which has been incorporated into the linker and attached to a modified nucleotide, is cleaved , a short “ scar ” or chemical modification on the nucleotide may result . For example, after cleaving , the resulting scar on a nucleotide base , which formed part of the modified nucleotide , and is incorporated into a polynucleotide strand , is unreactive and does not need to be chemically neutralized . This increases the ease with which a subsequent nucleotide can be incorporated during sequencing of a nucleic acid polymer template. For example , conditions include the use of tris ( 2 -carboxyethyl)phosphine ( TCEP ), dithiothreitol ( DTT ) and /or other reducing agents for cleavage of a disulfide bond . A selectively severable bond that includes an amido bond can be cleaved for example by the use of TCEP or other reducing agents, and / or photolysis. A selectively severable bond that includes an ester bond can be cleaved for example by acidic or basic hydrolysis. Detectable Agents The mmRNAs of the present invention may also be linked or conjugated to one or more detectable agents. Examples of detectable substances include , but are not limited to , various organic small molecules , inorganic compounds, nanoparticles , enzymes or enzyme substrates, fluorescent materials, luminescent materials, bioluminescent materials, chemiluminescent materials, radioactive materials , and contrast agents. 40 45 50 55 60 65 Endogenous eukaryotic cellular messenger RNA (mRNA ) molecules contain a 5 ' - cap structure on the 5 ' - end of a mature mRNA molecule . The 5 ' - cap contains a 5'- 5'- triphos phate linkage between the 5 ' -most nucleotide and guanine nucleotide . The conjugated guanine nucleotide is methylated at the N7 position . Additional modifications include meth ylation of the ultimate and penultimate most 5 '-nucleotides on the 2 '-hydroxyl group. The 5 ' - cap structure is responsible for binding the mRNA Cap Binding Protein ( CBP ) , which is responsibility for mRNA stability in the cell and translation competency . Multiple distinct 5 '-cap structures can be used to generate the 5 ' - cap of a synthetic mRNA molecule . Many chemical cap analogs are used to co - transcriptionally cap a synthetic mRNA molecule . For example, the Anti-Reverse Cap Ana log (ARCA) cap contains a 5 '- 5 ' -triphosphate guanine- gua nine linkage where one guanine contains an N7 methyl group as well as a 3 ' - O -methyl group . While chemical cap analogs allow for the concomitant capping of an RNA molecule , up 20 % of transcripts remain uncapped and the synthetic cap analog is not identical to an endogenous 5 ' - cap structure of an authentic cellular mRNA . This may lead to reduced translationally -competency and reduced cellular stability . Synthetic mRNA molecules may also be capped post transcriptionally using enzymes responsible for generating a more authentic 5 '-cap structure . As used herein the phrase “ more authentic” refers to a feature that closely mirrors or mimics , either structurally or functionally an endogenous or wild type feature . More authentic 5 ' - cap structures of the present invention are those which , among other things , have enhanced binding of cap binding proteins , increased half life , reduced susceptibility to 5 ' endonucleases and / or reduced 5'decapping. For example, recombinant Vaccinia Virus Capping Enzyme and recombinant 2 - O -methyltrans ferase enzyme can create a canonical 5 '- 5 '-triphosphate linkage between the 5 ' -most nucleotide of an mRNA and a guanine nucleotide where the guanine contains an N7 meth ylation and the ultimate 5 ' - nucleotide contains a 2 '- O -methyl generating the Capl structure . This results in a cap with higher translational - competency and cellular stability and reduced activation of cellular pro - inflammatory cytokines . Because the synthetic mRNA is capped post-transcription ally, nearly 100 % of the mRNA molecules are capped in contrast to ~ 80% of synthetic mRNAs containing a chemical cap analog . Terminal Architecture Modifications: Poly -A Tails During RNA processing , a long chain of adenine nucleo tides (poly - A tail ) is normally added to a messenger RNA (mRNA ) molecules to increase the stability of the molecule . Immediately after transcription, the 3 ' end of the transcript is cleaved to free a 3 ' hydroxyl. Then poly -A polymerase adds a chain of adenine nucleotides to the RNA . The process, called polyadenylation, adds a poly - A tail that is between 100 and 250 residues long . It has been discovered that unique poly - A tail lengths provide certain advantages to the modified RNAs of the present invention . US 10,898,574 B2 13 Generally , the length of a poly -A tail of the present invention is greater than 30 nucleotides in length . In another embodiment, the poly - A tail is greater than 35 nucleotides in 14 such that the mmRNA is localized into one or more cells of the cell population and the recombinant polypeptide is translated in the cell from the nucleic acid . length . In another embodiment, the length is at least 40 An effective amount of the composition is provided based , nucleotides . In another embodiment, the length is at least 45 5 at least in part, on the target tissue , target cell type , means nucleotides . In another embodiment, the length is at least 55 of administration , physical characteristics of the nucleic acid nucleotides . In another embodiment, the length is at least 60 (e.g. , size , and extent of modified nucleosides ), and other nucleotides . In another embodiment, the length is at least 60 determinants. In general, an effective amount of the com nucleotides. In another embodiment, the length is at least 80 position provides efficient protein production in the cell , nucleotides. In another embodiment, the length is at least 90 10 preferably more efficient than a composition containing a nucleotides . In another embodiment, the length is at least corresponding unmodified nucleic acid . Increased efficiency 100 nucleotides . In another embodiment, the length is at may be demonstrated by increased cell transfection (i.e. , the least 120 nucleotides. In another embodiment, the length is at least 140 nucleotides. In another embodiment, the length is at least 160 nucleotides . In another embodiment, the 15 length is at least 180 nucleotides . In another embodiment, the length is at least 200 nucleotides. In another embodiment, the length is at least 250 nucleotides . In another embodiment, the length is at least 300 nucleotides . In another embodiment, the length is at least 350 nucleotides. 20 In another embodiment, the length is at least 400 nucleotides . In another embodiment, the length is at least 450 nucleotides . In another embodiment, the length is at least 500 nucleotides. In another embodiment, the length is at least 600 nucleotides. In another embodiment, the length is 25 at least 700 nucleotides . In another embodiment, the length is at least 800 nucleotides. In another embodiment, the length is at least 900 nucleotides. In another embodiment, the length is at least 1000 nucleotides. In one embodiment, the poly -A tail is designed relative to 30 the length of the overall modified RNA molecule . This design may be based on the length of the coding region of the modified RNA , the length of a particular feature or region of the modified RNA ( such as the mRNA ), or based on the length of the ultimate product expressed from the 35 modified RNA . In this context the poly -A tail may be 10 , 20 , 30 , 40 , 50 , 60 , 70 , 80 , 90 or 100% greater in length than the modified RNA or feature thereof. The poly -A tail may also be designed as a fraction of the modified RNA to which it belongs. In this context, the poly -A tail may be 10 , 20 , 30 , 40 40 , 50 , 60 , 70 , 80 , or 90 % or more of the total length of the construct or the total length of the construct minus the percentage of cells transfected with the nucleic acid ), increased protein translation from the nucleic acid , decreased nucleic acid degradation (as demonstrated , e.g. , by increased duration of protein translation from a mmRNA ), or reduced innate immune response of the host cell . Aspects of the invention are directed to methods of inducing in vivo translation of a recombinant polypeptide in a mammalian subject in need thereof. Therein , an effective amount of a composition containing a mmRNA that has at least one nucleoside modification and a translatable region encoding the recombinant polypeptide is administered to the subject using the delivery methods and split dosing regimens described herein . The mmRNA is provided in an amount and under other conditions such that the nucleic acid is localized into a cell of the subject and the recombinant polypeptide is and in kits and devices. production of one or more recombinant polypeptides that translated in the cell from the mmRNA . The cell in which the mmRNA is localized , or the tissue in which the cell is present, may be targeted with one or more than one rounds of mmRNA administration . The subject to whom the therapeutic agent is administered suffers from or is at risk of developing a disease , disorder, or deleterious condition . Provided are methods of identifying, diagnosing, and classifying subjects on these bases , which may include clinical diagnosis , biomarker levels , genome wide association studies (GWAS ), and other methods known in the art. In certain embodiments , the administered mmRNA directs production of one or more recombinant polypeptides that provide a functional activity which is substantially poly -A tail . absent in the cell in which the recombinant polypeptide is Use of Modified mRNAS translated . For example, the missing functional activity may The mmRNAs of the present invention may find uses in 45 be enzymatic, structural, or gene regulatory in nature. In many areas of research , discovery, therapeutics, diagnostics related embodiments , the administered mmRNA directs Therapeutics increases ( e.g. , synergistically ) a functional activity which is The mmRNAs (modified RNAs) and the proteins trans- present but substantially deficient in the cell in which the lated from the mmRNAs described herein can be used as 50 recombinant polypeptide is translated . therapeutic agents. For example, an mmRNA described In other embodiments, the administered mmRNA directs herein can be administered to a subject, wherein the production of one or more recombinant polypeptides that mmRNA is translated in vivo to produce a therapeutic replace a polypeptide (or multiple polypeptides ) that is polypeptide in the subject. Provided are compositions, meth- substantially absent in the cell in which the recombinant ods , kits, and reagents for treatment or prevention of disease 55 polypeptide is translated. Such absence may be due to or conditions in humans and other mammals . The active therapeutic agents of the invention include mmRNAs, cells containing mmRNAs or polypeptides translated from the genetic mutation of the encoding gene or regulatory pathway thereof. In some embodiments, the recombinant polypeptide increases the level of an endogenous protein in the cell to a desirable level ; such an increase may bring the level of the mmRNAs, polypeptides translated from mmRNAs. Provided herein are methods of inducing translation of a 60 endogenous protein from a subnormal level to a normal level recombinant polypeptide in a cell population using the or from a normal level to a super -normal level. mmRNAs described herein . Such translation can be in vivo , Alternatively, the recombinant polypeptide functions to ex vivo , in culture , or in vitro . The cell population is antagonize the activity of an endogenous protein present in , contacted with an effective amount of a composition con- on the surface of, or secreted from the cell . Usually, the taining a mmRNA that has at least one nucleoside modifi- 65 activity of the endogenous protein is deleterious to the cation , and a translatable region encoding the recombinant subject; for example, do to mutation of the endogenous polypeptide . The population is contacted under conditions protein resulting in altered activity or localization . Addition US 10,898,574 B2 15 16 ally, the recombinant polypeptide antagonizes, directly or indirectly, the activity of a biological moiety present in , on the surface of, or secreted from the cell . Examples of antagonized biological moieties include lipids ( e.g. , choles- (e.g. , TLR7 and TLR8 ) . Reduction of innate immune response can also be measured by decreased cell death following one or more administrations of modified RNAs to a cell population ; e.g. , cell death is 10% , 25 % , 50% , 75 % , terol), a lipoprotein (e.g. , low density lipoprotein ), a nucleic 5 85 % , 90 % , 95 % , or over 95 % less than the cell death acid, a carbohydrate, a protein toxin such as shiga and frequency observed with a corresponding unmodified tetanus toxins , or a small molecule toxin such as botulinum , nucleic acid . Moreover, cell death may affect fewer than cholera, and diphtheria toxins. Additionally , the antagonized 50% , 40% , 30 % , 20 % , 10 % , 5 % , 1 % , 0.1 % , 0.01 % or fewer biological molecule may be an endogenous protein that than 0.01 % of cells contacted with the mmRNAs. exhibits an undesirable activity, such as a cytotoxic or 10 The invention provides therapeutic methods for the cytostatic activity. repeated introduction ( e.g. , transfection ) of mmRNAs into a The polypeptides encoded by the mmRNA described target cell population , e.g. , in vitro, ex vivo , or in vivo . The herein are engineered for localization within the cell , poten- step of contacting the cell population may be repeated one tially within a specific compartment such as the nucleus, or or more times ( such as two, three, four, five or more than five are engineered for secretion from the cell or translocation to 15 times ) . In some embodiments, the step of contacting the cell the plasma membrane of the cell . population with the mmRNAs is repeated a number of times In one embodiment of the invention are bifunctional sufficient such that a predetermined efficiency of protein mmRNA . As the name implies, bifunctional mmRNA are translation in the cell population is achieved . Given the reduced cytotoxicity of the target cell population provided those having or capable of at least two functions. The multiple functionalities of bifunctional mmRNAs 20 by the nucleic acid modifications, such repeated transfec may be encoded by the mRNA ( the function may not tions are achievable in a diverse array of cell types. manifest until the encoded product is translated ) or may be Protein Production a property of the RNA itself. It may be structural or The methods provided herein are useful for enhancing chemical. Bifunctional modified RNAs may comprise a protein product yield in a cell culture process. In a cell function that is covalently associated with the RNA or 25 culture containing a plurality of host cells , introduction of electrostatically associated. the modified mRNAs described herein results in increased In some embodiments , modified mRNAs and their protein production efficiency relative to a corresponding encoded polypeptides in accordance with the present inven- unmodified nucleic acid . Such increased protein production tion may be used for treatment of any of a variety of efficiency can be demonstrated , e.g. , by showing increased diseases , disorders, and / or conditions , including but not 30 cell transfection , increased protein translation from the limited to one or more of the following: autoimmune dis- nucleic acid , decreased nucleic acid degradation, and / or orders ( e.g. diabetes, lupus, multiple sclerosis, psoriasis, reduced innate immune response of the host cell . Protein rheumatoid arthritis ); inflammatory disorders (e.g. arthritis, pelvic inflammatory disease) ; infectious diseases (e.g. viral infections (e.g. , HIV , HCV, RSV) , bacterial infections, fungal infections, sepsis ) ; neurological disorders (e.g. Alzheimer's disease , Huntington's disease ; autism ; Duchenne muscular dystrophy ); cardiovascular disorders ( e.g. atherosclerosis, hypercholesterolemia , thrombosis, clotting disorders, angiogenic disorders such as macular degeneration ); proliferative disorders (e.g. cancer, benign neoplasms ) ; respiratory disorders (e.g. chronic obstructive pulmonary disease ) ; digestive disorders (e.g. inflammatory bowel disease, ulcers ) ; musculoskeletal disorders (e.g. fibromyalgia , arthritis ); endocrine , metabolic , and nutritional disorders (e.g. diabetes , osteoporosis ) ; urological disorders ( e.g. renal disease) ; psychological disorders (e.g. depression , schizophrenia ); skin disorders ( e.g. wounds, eczema ); blood and lymphatic disorders ( e.g. anemia , hemophilia ); etc. Avoidance of the Innate Immune Response The term “ innate immune response” includes a cellular response to exogenous single stranded nucleic acids , generally of viral or bacterial origin , which involves the induction of cytokine expression and release , particularly the interferons, and cell death . Protein synthesis is also reduced during the innate cellular immune response . While it is advantageous to eliminate the innate immune response in a cell , the invention provides modified mRNAs that substantially reduce the immune response , including interferon production can be measured by EI A , and protein activity can be measured by various functional assays known in the 35 art. The protein production may be generated in a continuous or a fed -batch mammalian process . Additionally, it is useful to optimize the expression of a specific polypeptide in a cell line or collection of cell lines of potential interest, particularly an engineered protein such 40 as a protein variant of a reference protein having a known activity. In one embodiment, provided is a method of opti mizing expression of an engineered protein in a target cell , by providing a plurality of target cell types , and indepen dently contacting with each of the plurality of target cell 45 types a modified mRNA encoding an engineered polypep tide. Additionally, culture conditions may be altered to increase protein production efficiency. Subsequently, the presence and / or level of the engineered polypeptide in the plurality of target cell types is detected and / or quantitated , 50 allowing for the optimization of an engineered polypeptide's expression by selection of an efficient target cell and cell culture conditions relating thereto . Such methods are par ticularly useful when the engineered polypeptide contains one or more post - translational modifications or has substan 55 tial tertiary structure , situations which often complicate efficient protein production . Gene Silencing The modified mRNAs described herein are useful to silence (i.e. , prevent or substantially reduce ) expression of signaling, without entirely eliminating such a response . In 60 one or more target genes in a cell population. A modified some embodiments, the immune response is reduced by mRNA encoding a polypeptide capable of directing 10 % , 20 % , 30 % , 40 % , 50 % , 50 % , 70% , 80% , 90% , 95 % , sequence - specific histone H3 methylation is introduced into 99 % , 99.9 % , or greater than 99.9 % as compared to the the cells in the population under conditions such that the immune response induced by a corresponding unmodified polypeptide is translated and reduces gene transcription of a nucleic acid . Such a reduction can be measured by expres- 65 target gene via histone H3 methylation and subsequent sion or activity level of Type 1 interferons or the expression heterochromatin formation . In some embodiments, the of interferon - regulated genes such as the toll - like receptors silencing mechanism is performed on a cell population US 10,898,574 B2 17 18 present in a mammalian subject. By way of non- limiting ways that modulate cell fate determination . Such agoniza example , a useful target gene is a mutated Janus Kinase - 2 tion is reversible or, alternatively, irreversible . family member, wherein the mammalian subject expresses Cellular Nucleic Acid Delivery the mutant target gene suffers from a myeloproliferative Methods of the present invention enhance nucleic acid 5 delivery into a cell population, in vivo , ex vivo , or in culture . disease resulting from aberrant kinase activity. Co - administration of modified mRNAs and siRNAs are For example, a cell culture containing a plurality of host ( e.g. , eukaryotic cells such as yeast or mammalian also provided herein . As demonstrated in yeast, sequence cells specific trans silencing is an effective mechanism for alter cells ) is contacted with a composition that contains an ing cell function . Fission yeast require two RNAi complexes enhanced nucleic acid having at least one nucleoside modi for siRNA -mediated heterochromatin assembly: the RNA- 10 fication , optionally , a translatable regionreagent . The orcompo sition alsoandgenerally contains a transfection other induced transcriptional silencing (RITS ) complex and the compound increases the efficiency of enhanced nucleic RNA -directed RNA polymerase complex ( RDRC ) (Mot acid uptakethat into the host cells . The enhanced nucleic acid amedi et al . Cell 2004 , 119 , 789-802 ) . In fission yeast , the exhibits enhanced retention in the cell population , relative to RITS complex contains the siRNA binding Argonaute fam15 a corresponding unmodified nucleicacid. The retention of ily protein Agol, a chromodomain protein Chpl, and Tas3 . the enhanced nucleic acid is greater than the retention of the The fission yeast RDRC complex is composed of an RNA unmodified nucleic acid . In some embodiments least dependent RNA Polymerase Rdp1, a putative RNA helicase about 50% , 75 % , 90 % , 95 % , 100 % , 150 % , 200, it%isoratmore Hrr1, and a polyA polymerase family protein Cid12 . These than 200 % greater than the retention of the unmodified two complexes require the Dicer ribonuclease and Clr4 20 nucleic acid. Such retention advantage may be achieved by histone H3 methyltransferase for activity . Together, Ago 1 one round of transfection with the enhanced nucleic acid, or binds siRNA molecules generated through Dicer-mediated may be obtained following repeated rounds of transfection . cleavage of Rdpl co - transcriptionally generated dsRNA In some embodiments , the enhanced nucleic acid is deliv transcripts and allows for the sequence - specific direct asso- ered to a target cell population with one or more additional ciation of Chpl, Tas3 , Hrrl, and Clr4 to regions of DNA 25 nucleic acids . Such delivery may be at the same time , or the destined for methylation and histone modification and sub- enhanced nucleic acid is delivered prior to delivery of the sequent compaction into transcriptionally silenced heterochromatin . While this mechanism functions in cis- with centromeric regions of DNA , sequence -specific trans silenc- one or more additional nucleic acids . The additional one or more nucleic acids may be modified nucleic acids or unmodified nucleic acids . It is understood that the initial ing is possible through co - transfection with double - stranded 30 presence of the enhanced nucleic acids does not substan siRNAs for specific regions of DNA and concomitant RNAi- tially induce an innate immune response of the cell popu directed silencing of the siRNA ribonuclease Eril (Buhler et al . Cell 2006 , 125 , 873-886 ) . Modulation of Biological Pathways lation and, moreover, that the innate immune response will not be activated by the later presence of the unmodified nucleic acids . In this regard , the enhanced nucleic acid may The rapid translation of modified mRNAs introduced into 35 not itself contain a translatable region , if the protein desired cells provides a desirable mechanism of modulating target to be present in the target cell population is translated from biological pathways. Such modulation includes antagonism the unmodified nucleic acids . or agonism of a given pathway. In one embodiment, a Expression of Ligand or Receptor on Cell Surface method is provided for antagonizing a biological pathway in In some aspects and embodiments of the aspects a cell by contacting the cell with an effective amount of a 40 described herein , the modified RNAs can be used to express composition comprising a modified nucleic acid encoding a a ligand or ligand receptor on the surface of a cell (e.g. , a recombinant polypeptide , under conditions such that the homing moiety ). A ligand or ligand receptor moiety attached nucleic acid is localized into the cell and the recombinant to a cell surface can permit the cell to have a desired polypeptide is capable of being translated in the cell from the biological interaction with a tissue or an agent in vivo . A nucleic acid, wherein the recombinant polypeptide inhibits 45 ligand can be an antibody, an antibody fragment, an aptamer, the activity of a polypeptide functional in the biological a peptide , a vitamin , a carbohydrate, a protein or polypep pathway. Exemplary biological pathways are those defective tide, a receptor, e.g. , cell- surface receptor, an adhesion in an autoimmune or inflammatory disorder such as multiple molecule, a glycoprotein , a sugar residue, a therapeutic sclerosis, rheumatoid arthritis, psoriasis , lupus erythemato- agent, a drug, a glycosaminoglycan, or any combination sus , ankylosing spondylitis colitis , or Crohn's disease; in 50 thereof. For example , a ligand can be an antibody that particular, antagonism of the IL - 12 and IL - 23 signaling recognizes a cancer- cell specific antigen, rendering the cell pathways are of particular utility . ( See Kikly K , Liu L , Na capable of preferentially interacting with tumor cells to S , Sedgwick J D (2006 ) Curr. Opin . Immunol. 18 ( 6 ) : permit tumor -specific localization of a modified cell . A 670-5) . ligand can confer the ability of a cell composition to Further, provided are modified nucleic acids encoding an 55 accumulate in a tissue to be treated , since a preferred ligand antagonist for chemokine receptors ; chemokine receptors may be capable of interacting with a target molecule on the CXCR - 4 and CCR- 5 are required for, e.g. , HIV entry into external face of a tissue to be treated . Ligands having limited cross -reactivity to other tissues are generally preferred . host cells (et al , ( 1996 ) October 3 ; 383 ( 6599 ) : 400 ) . Alternatively, provided are methods of agonizing a bioIn some cases , a ligand can act as a homing moiety which logical pathway in a cell by contacting the cell with an 60 permits the cell to target to a specific tissue or interact with effective amount of a modified nucleic acid encoding a a specific ligand. Such homing moieties can include , but are recombinant polypeptide under conditions such that the not limited to , any member of a specific binding pair, nucleic acid is localized into the cell and the recombinant antibodies , monoclonal antibodies, or derivatives or analogs polypeptide is capable of being translated in the cell from the thereof, including without limitation : Fv fragments, single nucleic acid, and the recombinant polypeptide induces the 65 chain Fv ( scFv) fragments, Fab ' fragments , F ( ab ' )2 frag activity of a polypeptide functional in the biological pathway. Exemplary agonized biological pathways include path- ments , single domain antibodies, camelized antibodies and antibody fragments, humanized antibodies and antibody US 10,898,574 B2 19 20 fragments, and multivalent versions of the foregoing; mul- totic when formed into trimers or higher order of structures. tivalent binding reagents including without limitation : As monomers , they may serve as antiapoptotic agents by monospecific or bispecific antibodies, such as disulfide competing with the trimers for binding to the death recep stabilized Fv fragments, scFv tandems ( ( SCFV) 2 frag- tors . ments ), diabodies , tribodies or tetrabodies, which typically 5 In one embodiment, the modified nucleic acid molecule are covalently linked or otherwise stabilized ( i.e. , leucine composition encodes for a death receptor (e.g. , Fas , TRAIL , zipper or helix stabilized ) scFv fragments; and other homing TRAMO , TNFR , TLR etc ). Cells made to express a death moieties include for example, aptamers, receptors, and receptor by transfection of modified RNA become suscep fusion proteins. tible to death induced by the ligand that activates that In some embodiments, the homing moiety may be a 10 receptor. Similarly, cells made to express a death ligand, e.g. , surface - bound antibody, which can permit tuning of cell on their surface, will induce death of cells with the receptor targeting specificity. This is especially useful since highly when the transfected cell contacts the target cell . In another specific antibodies can be raised against an epitope of embodiment, the modified RNA composition encodes for a interest for the desired targeting site . In one embodiment, death receptor ligand (e.g. , FasL , TNF, etc ) . In another multiple antibodies are expressed on the surface of a cell , 15 embodiment, the modified RNA composition encodes a and each antibody can have a different specificity for a caspase ( e.g. , caspase 3 , caspase 8 , caspase 9 etc ). Where desired target. Such approaches can increase the avidity and cancer cells often exhibit a failure to properly differentiate to specificity of homing interactions . a non - proliferative or controlled proliferative form , in A skilled artisan can select any homing moiety based on another embodiment, the synthetic, modified RNA compo the desired localization or function of the cell , for example 20 sition encodes for both a death receptor and its appropriate an estrogen receptor ligand , such as tamoxifen , can target activating ligand. In another embodiment, the synthetic , cells to estrogen -dependent breast cancer cells that have an modified RNA composition encodes for a differentiation increased number of estrogen receptors on the cell surface . factor that when expressed in the cancer cell , such as a Other non - limiting examples of ligand/ receptor interactions cancer stem cell , will induce the cell to differentiate to a include CCRI (e.g. , for treatment of inflamed joint tissues or 25 non - pathogenic or nonself- renewing phenotype ( e.g. , brain in rheumatoid arthritis , and /or multiple sclerosis ), CCR7 , CCR8 ( e.g. , targeting to lymph node tissue ) , CCR6 , CCR9 , CCR10 (e.g. , to target to intestinal tissue ) , CCR4 , reduced cell growth rate , reduced cell division etc ) or to induce the cell to enter a dormant cell phase (e.g. , G. resting phase ). CCR10 ( e.g. , for targeting to skin) , CXCR4 (e.g. , for general One of skill in the art will appreciate that the use of enhanced transmigration ), HCELL ( e.g. , for treatment of 30 apoptosis - inducing techniques may require that the modified inflammation and inflammatory disorders, bone marrow ), nucleic acid molecules are appropriately targeted to e.g. , Alpha4beta 7 (e.g. , for intestinal mucosa targeting ), VLA - 4 / tumor cells to prevent unwanted wide - spread cell death . VCAM - 1 ( e.g. , targeting to endothelium ). In general, any hus, one can use a delivery mechanism ( e.g. , attached receptor involved in targeting ( e.g. , cancer metastasis ) can ligand or antibody, targeted liposome etc) that recognizes a be harnessed for use in the methods and compositions 35 cancer antigen such that the modified nucleic acid molecules described herein . are expressed only in cancer cells . Mediators of Cell Death Formulations of Modified mRNAS In one embodiment, a modified nucleic acid molecule Provided herein are formulations containing an effective composition can be used to induce apoptosis in a cell ( e.g. , amount of an mmRNA . a cancer cell ) by increasing the expression of a death 40 In certain embodiments, the formulations include one or receptor, a death receptor ligand or a combination thereof. more cell penetration agents, e.g. , transfection agents . In one This method can be used to induce cell death in any desired specific embodiment, an mmRNA is mixed or admixed with cell and has particular usefulness in the treatment of cancer a transfection agent ( or mixture thereof) and the resulting where cells escape natural apoptotic signals . mixture is employed to transfect cells . Preferred transfection Apoptosis can be induced by multiple independent sig- 45 agents are cationic lipid compositions, particularly monova naling pathways that converge upon a final effector mecha- lent and polyvalent cationic lipid compositions, more par nism consisting of multiple interactions between several ticularly LIPOFECTIN® , LIPOFECTACE® , LIPO “ death receptors ” and their ligands, which belong to the FECTAMINETM , CELLFECTIN® , DMRIE - C , DMRIE , tumor necrosis factor ( TNF) receptor /ligand superfamily. DOTAP, DOSPA , and DOSPER , and dendrimer composi The best - characterized death receptors are CD95 (“ Fas ” ), 50 tions , particularly G5 - G10 dendrimers, including dense star TNFRI ( p55 ), death receptor 3 (DR3 or Apo3 / TRAMO ), dendrimers, PAMAM dendrimers , grafted dendrimers, and DR4 and DR5 (apo 2 -TRAIL -R2). The final effector mecha- dendrimers known as dendrigrafts and SUPERFECT® . nism of apoptosis may be the activation of a series of In a second specific transfection method, a ribonucleic proteinases designated as caspases . The activation of these acid is conjugated to a nucleic acid -binding group , for caspases results in the cleavage of a series of vital cellular 55 example a polyamine and more particularly a spermine, proteins and cell death . The molecular mechanism of death which is then introduced into the cell or admixed with a receptors / ligands -induced apoptosis is well known in the art. transfection agent ( or mixture thereof) and the resulting For example, Fas /FasL - mediated apoptosis is induced by mixture is employed to transfect cells . In a third specific binding of three Fasl molecules which induces trimerization embodiment, a mixture of one or more transfection - enhanc of Fas receptor via C - terminus death domains ( DDs ) , which 60 ing peptides, proteins , or protein fragments, including fus in turn recruits an adapter protein FADD (Fas - associated agenic peptides or proteins, transport or trafficking peptides protein with death domain) and Caspase - 8 . The oligomer- or proteins, receptor- ligand peptides or proteins , or nuclear ization of this trimolecular complex , Fas /FAIDD /caspase - 8, localization peptides or proteins and / or their modified ana results in proteolytic cleavage of proenzyme caspase - 8 into logs (e.g. , spermine modified peptides or proteins) or com active caspase - 8 that, in turn , initiates the apoptosis process 65 binations thereof are mixed with and complexed with a by activating other downstream caspases through proteoly- ribonucleic acid to be introduced into a cell , optionally being sis , including caspase - 3 . Death ligands in general are apop- admixed with transfection agent and the resulting mixture is US 10,898,574 B2 21 22 employed to transfect cells . Further, a component of a According to the present invention , complexes , micelles, In other embodiments, provided are sustained -release delivery depots , such as for administration of a mmRNA to is not limited to , DLin - DMA , DLin - K - DMA, DLin -KC2 DMA , 12-5 , C12-200 ( including variants and deriva transfection agent (e.g. , lipids , cationic lipids or dendrimers ) liposomes or particles can be prepared containing these is covalently conjugated to selected peptides , proteins , or lipidoids and therefore, result in an effective delivery of protein fragments directly or via a linking or spacer group . mmRNA, as judged by the production of an encoded pro Of particular interest in this embodiment are peptides or 5 tein , following the injection of an mmRNA - formulated proteins that are fusagenic, membrane -permeabilizing, lipidoids via localized and systemic routes of administration . transport or trafficking, or which function for cell- targeting. Modified mRNA - lipidoid complexes can be administered by The peptide- or protein -transfection agent complex is com- various means disclosed herein . bined with a ribonucleic acid and employed for transfection . The characteristics of optimized lipidoid formulations for In certain embodiments, the formulations include a phar- 10 intramuscular or subcutaneous routes may vary significantly maceutically acceptable carrier that causes the effective depending on the target cell type and the ability of formu amount of mmRNA to be substantially retained in a target lations to diffuse through the extracellular matrix into the tissue containing the cell . blood stream . While a particle size of less than 150 nm may In certain embodiments, the formulation may include at be desired for effective hepatocyte delivery due to the size least an mmRNA and a delivery agent. In some embodi- 15 of the endothelial fenestrae ( see , Akinc et al . , Mol Ther. ments, the delivery agent may comprise lipidoid -based 2009 17 : 872-879 herein incorporated by reference ), use of formulations allowed for localized and systemic delivery of lipidoid oligonucleotides to deliver the formulation to other mmRNA . cells types including , but not limited to , endothelial cells , Also provided are compositions for generation of an in myeloid cells , and muscle cells may not be similarly size vivo depot containing an engineered ribonucleotide . For 20 limited . example , the composition contains a bioerodible , biocomIn one aspect , effective delivery to myeloid cells , such as patible polymer, a solvent present in an amount effective to monocytes, lipidoid formulations may have a similar com plasticize the polymer and form a gel therewith , and an ponent molar ratio . Different ratios of lipidoids and other engineered ribonucleic acid . In certain embodiments the components including , but not limited to , disteroylphospha composition also includes a cell penetration agent as 25 tidyl choline , cholesterol and PEG - DMG , may be used to described herein . In other embodiments, the composition optimize the formulation of the mmRNA molecule for also contains a thixotropic amount of a thixotropic agent delivery to different cell types including , but not limited to , mixable with the polymer so as to be effective to form a hepatocytes, myeloid cells , muscle cells , etc. For example, thixotropic composition. Further compositions include a the component molar ratio may include, but is not limited to , stabilizing agent, a bulking agent, a chelating agent, or a 30 50% lipid, 10% disteroylphosphatidyl choline, 38.5 % cho buffering agent. lesterol, and % 1.5 PEG . The lipid may be selected from , but an environment (meaning an organ or tissue site) in a patient. Such depots generally contain a mmRNA and a flexible 35 chain polymer where both the mmRNA and the flexible chain polymer are entrapped within a porous matrix of a crosslinked matrix protein. Usually, the pore size is less than 1 mm , such as 900 nm , 800 nm , 700 nm , 600 nm , 500 nm , 400 nm , 300 nm , 200 nm , 100 nm , or less than 100 nm . 40 Usually the flexible chain polymer is hydrophilic. Usually the flexible chain polymer has a molecular weight of at least 50 kDa , such as 75 kDa , 100 kDa , 150 kDa , 200 kDa, 250 kDa , 300 kDa , 400 kDa, 500 kDa, or greater than 500 kDa . Usually the flexible chain polymer has a persistence length 45 of less than 10 % , such as 9 , 8 , 7 , 6 , 5 , 4 , 3 , 2 , 1 or less than 1 % of the persistence length of the matrix protein . Usually the flexible chain polymer has a charge similar to that of the matrix protein . In some embodiments, the flexible chain polymer alters the effective pore size of a matrix of cross- 50 linked matrix protein to a size capable of sustaining the diffusion of the mmRNA from the matrix into a surrounding tissue comprising a cell into which the mmRNA is capable of entering Formulation Using Lipidoids 55 The pharmaceutical compositions described herein include lipidoid -based formulations allowing for localized and systemic delivery of mmRNA . The synthesis of lipidoids has been extensively described and formulations con- tives ) , DLin -MC3 - DMA and analogs thereof. The use of lipidoid formulations for the localized delivery of nucleic acids to cells ( such as , but not limited to , adipose cells and muscle cells ) via either subcutaneous or intramuscular deliv ery , may also not require all of the formulation components which may be required for systemic delivery, and as such may comprise the lipidoid and the mmRNA . In a further embodiment, combinations of different lipi doids may be used to improve the efficacy of mmRNA directed protein. According to the present invention , modified mRNA may be formulated by mixing the mmRNA with the lipidoid at a set ratio prior to addition to cells . In vivo formulations may require the addition of extra ingredients to facilitate circu lation throughout the body. To test the ability of these lipidoids to form particles suitable for in vivo work , a standard formulation process used for siRNA - lipidoid for mulations may be used as a starting point. Initial mmRNA lipidoid formulations consist of particles composed of 42 % lipidoid, 48 % cholesterol and 10 % PEG , with further opti mization of ratios possible . After formation of the particle , In vivo delivery of nucleic acids may be affected by many parameters, including, but not limited to , the formulation 21 : 1448-1454 ; Schroeder et al . , J Intern Med . 2010 267: 921 ; Akinc et al . , Nat Biotechnol. 2008 26 : 561-569 ; Love et eters such as particle size (Akinc et al . , Mol Ther. 2009 17 : 872-879 ; herein incorporated by reference in its entirety ). mmRNA is added and allowed to integrate with the com plex . The encapsulation efficiency is determined using a standard dye exclusion assays . taining these compounds are particularly suited for delivery 60 composition, nature of particle PEGylation, degree of load of polynucleotides ( see Mahon et al . , Bioconjug Chem . 2010 ing , oligonucleotide to lipid ratio , and biophysical param al . , Proc Natl Acad Sci USA . 2010 107 : 1864-1869 ; Siegwart As an example, small changes in the anchor chain length of which are incorporated herein by reference in their entire- effects on in vivo efficacy. Formulations with the different lipidoids , including , but not limited to penta [ 3-( 1 -laurylami et al . , Proc Natl Acad Sci USA . 2011 108 :12996-3001 ; all of 65 poly ( ethylene glycol) (PEG ) lipids may result in significant ties) . US 10,898,574 B2 23 24 nopropionyl) ] - triethylenetetramine hydrochloride ( TETA5LAP ; aka 98N12-5 , see Murugaiah et al . , Analytical Biochemistry, 401 : 61 ( 2010 ) ) , C12-200 ( including derivatives and variants ), MD1 , DLin - DMA , DLin - K - DMA, DLin- depending on the body weight of the animal . At various points in time points following the administration of the mmRNA - lipidoid , serum , tissues , and tissue lysates can be obtained and the level of the mmRNA -encoded product KC2 - DMA and DLin -MC3 - DMA ( see FIG . 1 ) , can be tested 5 determined . The ability of lipidoid -formulated mmRNA to for in vivo activity. express the desired protein product can be confirmed by The lipidoid referred to herein as “ 98N12-5 ” is disclosed luminescence for luciferase expression , flow cytometry, and by Akinc et al . , Mol Ther. 2009 17 : 872-879 and is incor- by ELISA . or more components in addition to polynucleotide, primary construct, or mmRNA. As an example, formulations with then determining whether expression level is affected by factors such as immunogenicity ). porated by reference in its entirety. ( See FIG . 1 ) Additional studies for a multi -dose regimen can also be The lipidoid referred to herein as “ C12-200 ” is disclosed 10 performed to determine the maximal expression using by Love et al . , Proc Natl Acad Sci USA . 2010 107 : 1864- mmRNA , to evaluate the saturability of the mmRNA - driven 1869 ( see FIG . 1 ) and Liu and Huang, Molecular Therapy. expression (achieved by giving a control and active 2010 669-670 ( see FIG . 1 ) ; both of which are herein mmRNA formulation in parallel or in sequence ), and to incorporated by reference in their entirety. The lipidoid determine the feasibility of repeat drug administration (by formulations can include particles comprising either 3 or 4 15 giving mmRNA in doses separated by weeks or months and certain lipidoids, include, but are not limited to , 98N12-5 and may contain 42 % lipidoid , 48 % cholesterol and 10 % Administration The present invention provides methods comprising lations with certain lipidoids , include , but are not limited to , C12-200 and may contain 50% lipidoid , 10 % disteroylphosphatidyl choline , 38.5 % cholesterol, and 1.5 % PEG - DMG . The ratio of mmRNA to lipidoid used to test for in vitro transfection is tested empirically at different lipidoid : 25 mmRNA ratios. Previous work using siRNA and lipidoids have utilized 2.5 : 1 , 5 : 1 , 10 : 1 , and 15 : 1 lipidoid :siRNA wt:wt ratios . Given the longer length of mmRNA relative to siRNA , a lower wt : wt ratio of lipidoid to mmRNA is likely to be effective. In addition, for comparison mmRNA are also 30 formulated using RNAiMax ( Invitrogen, Carlsbad, Calif.) or TRANSIT -mRNA (Mirus Bio , Madison Wis .) cationic lipid delivery vehicles. The ability of lipidoid - formulated mmRNA to express the desired protein product can be confirmed by luminescence 35 for luciferase expression, flow cytometry for expression, and by ELISA for secretion . The expression of mmRNA - encoded proteins can be assessed both within the muscle or subcutaneous tissue and systemically in blood and other organs and fluids such as the 40 liver and spleen, urine, saliva, etc. For example, single dose studies allow an assessment of the magnitude, dose responsiveness, and longevity of expression of the desired product. After formulation of mmRNA with the lipidoid formulations, as described pre- 45 viously, animals are divided into groups receiving either a saline formulation, or a lipidoid - formulation containing one of several different mmRNA . Prior to injection , mmRNAcontaining lipidoid formulations are diluted in PBS and dance with the invention to a subject in need thereof. mmRNA or complexes, or pharmaceutical, imaging , diag nostic , or prophylactic compositions thereof, may be admin istered to a subject using any amount and any route of administration which may be effective for preventing, treat ing , diagnosing, or imaging a disease , disorder, and / or condition (e.g. , a disease , disorder, and / or condition relating PEG (C14 alkyl chain length ). As another example, formu- 20 administering modified mRNAs and or complexes in accor to working memory deficits ). The exact amount required will vary from subject to subject, depending on factors such as , but not limited to , the species , age , and general condition of the subject, the severity of the disease , the particular composition, its mode of administration , its mode of activity, and the like . mmRNA to be delivered and / or pharmaceutical, prophy lactic , diagnostic, or imaging compositions thereof may be administered to animals, such as mammals (e.g. , humans, domesticated animals, cats , dogs , mice , rats , etc. ) . In some embodiments , pharmaceutical, prophylactic , diagnostic , or imaging compositions thereof are administered to humans . mmRNA may be administered by any route . In some embodiments, mmRNA are administered by one or more of a variety of routes , including , but not limited to , local , oral , intravenous, intramuscular, intra - arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (e.g. by powders, ointments, creams, gels , lotions , and / or drops ), mucosal, nasal , buccal , enteral, vitreal, intratumoral, sublin gual ; by intratracheal instillation , bronchial instillation , and / or inhalation ; as an oral spray, nasal spray , and / or aerosol, animals administered a single intramuscular dose of formu- 50 and /or through a portal vein catheter. lated mmRNA ranging from 50 mg/kg to doses as low as 1 In some embodiments , mmRNA are administered by ng /kg with a preferred range to be 10 mg/kg to 100 ng /kg. systemic intravenous injection . In specific embodiments, If the animal tested is a mouse the maximum dose can be mmRNA may be administered intravenously and / or orally. roughly 1 mg mmRNA or as low as 0.02 ng mmRNA if In specific embodiments, mmRNA may be administered in administered once into the hind limb. Likewise for subcu- 55 a way which allows the mmRNA to cross the blood -brain taneous administration , mmRNA - containing lipidoid formu- barrier, vascular barrier, or other epithelial barrier. lations are diluted in PBS before the animals are adminisInjectable preparations, for example, sterile injectable tered a single subcutaneous dose of formulated mmRNA aqueous or oleaginous suspensions may be formulated ranging from 400 mg/kg- to doses as low as 1 ng /kg. A according to the known art using suitable dispersing agents, preferred dosage range comprises 80 mg /kg to 100 ng /kg. If 60 wetting agents, and / or suspending agents. Sterile injectable the animal tested is a mouse , the maximum dose adminis- preparations may be sterile injectable solutions , suspen tered can be roughly 8 mg mmRNA or as low as 0.02 ng sions , and / or emulsions in nontoxic parenterally acceptable mmRNA if the dose is administered once subcutaneously. diluents and / or solvents, for example, as a solution in ous injection is maximally 0.2 ml for a 20 gram mouse . The and isotonic sodium chloride solution . Sterile , fixed oils are It is preferred that the volume of a single intramuscular 1,3 - butanediol. Among the acceptable vehicles and solvents injection is maximally 0.025 ml and of a single subcutane- 65 that may be employed are water, Ringer's solution , U.S.P., dose of the mmRNA administered to the animal is calculated conventionally employed as a solvent or suspending US 10,898,574 B2 25 26 embodiments, the desired dosage may be delivered using medium . For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such. as oleic acid can be used in the preparation of injectables multiple administrations ( e.g. , two , three, four, five , six , seven , eight, nine , ten, eleven, twelve , thirteen , fourteen , or filtration through a bacterial - retaining filter, and / or by incor porating sterilizing agents in the form of sterile solid com positions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use . herein may be used . According to the present invention , it has been discovered that administration of mmRNA in split -dose regimens pro more administrations ). When multiple administration is Injectable formulations can be sterilized, for example, by 5 employed , split dosing regimens such as those described duce higher levels of proteins in mammalian subjects . As or total daily dose into two or more doses . As used herein , a “ single unit dose ” is a dose of any therapeutic administered Dosageofforms for local, topical and/or transdermal istration a composition may include ointments, adminpastes , 10 used herein, a “ split dose” is the division of single unit dose creams, lotions , gels , powders , solutions , sprays, inhalants and / or patches. Additionally, the present invention contem plates the use of transdermal patches, which often have the in one dose /at one time/ single route / single point of contact, added advantage of providing controlled delivery of a com- 15 i.e. , single administration event. As used herein, a “total pound to the body. Such dosage forms may be prepared, for daily dose” is an amount given or prescribed in 24 hr period . example, by dissolving and /or dispensing the compound in the proper medium . Alternatively or additionally , rate may be controlled by either providing a rate controlling membrane and / or by dispersing the compound in a polymer 20 matrix and /or gel . Formulations suitable for topical administration include, It may be administered as a single unit dose . In one embodiment, the mmRNA of the present invention are administered to a subject in split doses . The mmRNA may be formulated in buffer only or in a formulation described herein . Modified nucleic acid molecules or complexes may be mulations may, for example, comprise from about 1 % to about 10 % ( w / w ) active ingredient, although the concentra tion of active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for 30 topical administration may further comprise one or more of the additional ingredients described herein . A pharmaceutical composition may be prepared , pack aged , and / or sold in a formulation suitable for ophthalmic administration. Such formulations may, for example, be in 35 the form of eye drops including, for example, a 0.1 / 1.0 % ( w / w ) solution and / or suspension of the active ingredient in an aqueous or oily liquid excipient. Such drops may further mulated for delivery together, although these methods of delivery are within the scope of the present disclosure . Compositions can be administered concurrently with , prior to , or subsequent to , one or more other desired therapeutics or medical procedures. In general, each agent will be admin but are not limited to , liquid and / or semi liquid preparations used or administered in combination with one or more other such as liniments, lotions , oil in water and /or water in oil therapeutic , prophylactic, diagnostic, or imaging agents. By emulsions such as creams, ointments and / or pastes , and / or 25 “ in combination with ,” it is not intended to imply that the solutions and / or suspensions. Topically -administrable for- agents must be administered at the same time and / or for comprise buffering agents, salts , and / or one or more other of any additional ingredients described herein . Other opthal- 40 mically -administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form and / or in a liposomal preparation. Ear drops and / or eye drops are contemplated as being within the scope of this invention . 45 In general the most appropriate route of administration will depend upon a variety of factors including the nature of the mmRNA to be delivered (e.g. , its stability in the environment of the gastrointestinal tract, bloodstream , etc. ), the condition of the patient ( e.g. , whether the patient is able to 50 tolerate particular routes of administration ), etc. The invention encompasses the delivery of the mmRNA by any appropriate route taking into consideration likely advances in the sciences of drug delivery. In certain embodiments, compositions in accordance with 55 the present invention may be administered at dosage levels sufficient to deliver from about 0.0001 mg /kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg /kg, from about 0.1 mg/kg to about 40 mg /kg, from about 0.5 mg /kg to about 30 mg /kg, from about 0.01 mg /kg to about 10 60 mg/kg, from about 0.1 mg /kg to about 10 mg /kg, or from about 1 mg /kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired thera- istered at a dose and /or on a time schedule determined for that agent. In some embodiments, the present disclosure encompasses deliverycompositions of pharmaceutical , prophylactic diagnostic, ortheimaging in combination with, agents that may improve their bioavailability, reduce and /or modify their metabolism , inhibit their excretion, and / or modify their distribution within the body. It will further be appreciated that therapeutically, prophy lactically, diagnostically, or imaging active agents utilized in combination may be administered together in a single com position or administered separately in different composi tions . In general, it is expected that agents utilized in combination with be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually. In one embodiment, the combinations, each or together may be administered according to the split dosing regimens described herein . The particular combination of therapies ( therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and /or procedures and the desired therapeutic effect to be achieved . It will also be appreciated that the therapies employed may achieve a desired effect for the same disorder ( for example , a composition useful for treating cancer in accordance with the invention may be administered concurrently with a chemotherapeutic agent ), or they may achieve different effects (e.g. , control of any adverse effects ). Compositions containing mmRNAs are formulated for administration intramuscularly, transarterially , intraocularly, vaginally , rectally, intraperitoneally, intravenously, intrana peutic , diagnostic or prophylactic effect. The desired dosage sally, subcutaneously, endoscopically, transdermally, intra may be delivered three times a day, two times a day, once a 65 muscularly, intraventricularly, intradermally, intrathecally, day, every other day, every third day, every week, every two topically ( e.g. by powders, ointments , creams, gels , lotions , weeks, every three weeks , or every four weeks . In certain and / or drops), mucosally, nasal , enterally, intratumorally , by US 10,898,574 B2 27 28 intratracheal instillation , bronchial instillation , and / or inha- of interest in a substantial percentage of cells contained lation ; nasal spray and / or aerosol, and /or through a portal within a predetermined volume of the target tissue . In some vein catheter. embodiments, the composition includes a plurality of dif The compositions may also be formulated for direct ferent ribonucleic acids , where one or more than one of the delivery to an organ or tissue in any of several ways in the 5 ribonucleic acids is engineered to avoid an innate immune art including, but not limited to , direct soaking or bathing, response of a cell into which the ribonucleic acid enters , and via a catheter, by gels , powder, ointments , creams , gels , where one or more than one of the ribonucleic acids encodes lotions , and / or drops, by using substrates such as fabric or a polypeptide of interest. Optionally, the composition also biodegradable materials coated or impregnated with the contains a cell penetration agent to assist in the intracellular compositions , and the like . In some embodiments , the com- 10 delivery of the ribonucleic acid . A determination is made of position is formulated for extended release . In specific the dose of the composition required to produce the poly embodiments, mmRNA molecules or complexes, and / or peptide of interest in a substantial percentage of cells pharmaceutical, prophylactic , diagnostic , or imaging com- contained within the predetermined volume of the target positions thereof, may be administered in a way which tissue ( generally, without inducing significant production of allows the mmRNA molecules or complex to cross the 15 the polypeptide of interest in tissue adjacent to the prede blood- brain barrier, vascular barrier, or other epithelial bar- termined volume, or distally to the target tissue ) . Subsequent rier. to this determination , the determined dose is introduced In some aspects of the invention, the nucleic acids (par- directly into the tissue of the mammalian subject. ticularly ribonucleic acids encoding polypeptides) are spaFormulations which may be administered intramuscularly tially retained within or proximal to a target tissue . Provided 20 and / or subcutaneously may include , but are not limited to , are method of providing a composition to a target tissue of polymers, copolymers, and gels. The polymers, copolymers a mammalian subject by contacting the target tissue (which and / or gels may further be adjusted to modify release contains one or more target cells ) with the composition kinetics by adjusting factors such as , but not limited to , under conditions such that the composition, in particular the molecular weight, particle size , payload and / or ratio of the nucleic acid component ( s) of the composition, is substan- 25 monomers . As a non -limiting example, formulations admin tially retained in the target tissue , meaning that at least 10 , istered intramuscularly and / or subcutaneously may include a 20 , 30 , 40 , 50 , 60 , 70 , 80 , 85 , 90 , 95 , 96 , 97 , 98 , 99 , 99.9 , copolymer such as poly (lactic - co - glycolic acid) . 99.99 or greater than 99.99 % of the composition is retained Localized delivery of the compositions described herein in the target tissue . Advantageously, retention is determined may be administered by methods such as , but not limited to , by measuring the amount of the nucleic acid present in the 30 topical delivery, ocular delivery, transdermal delivery, and composition that enters one or more target cells . For the like . The composition may also be administered locally example , at least 1 , 5 , 10 , 20 , 30 , 40 , 50 , 60 , 70 , 80 , 85 , 90 , to a part of the body not normally available for localized 95 , 96 , 97 , 98 , 99 , 99.9 , 99.99 or greater than 99.99 % of the delivery such as , but limited to , when a subject's body nucleic acids administered to the subject are present intra- is open to the environment during treatment. The composi cellularly at a period of time following administration . For 35 tion may further be delivered by bathing, soaking and / or example , intramuscular injection to a mammalian subject is surrounding the body part with the composition. performed using an aqueous composition containing a riboHowever, the present disclosure encompasses the delivery nucleic acid and a transfection reagent, and retention of the of mmRNA molecules or complexes, and / or pharmaceutical, composition is determined by measuring the amount of the prophylactic , diagnostic , or imaging compositions thereof, 40 by any appropriate route taking into consideration likely ribonucleic acid present in the muscle cells . Aspects of the invention are directed to methods of advances in the sciences of drug delivery . providing a composition to a target tissue of a mammalian The level or concentration of a mmRNA may be charac subject, by contacting the target tissue (containing one or terized using exosomes . A level or concentration of the more target cells ) with the composition under conditions mmRNA in exosomes can represent an expression level , such that the composition is substantially retained in the 45 presence , absence , truncation or alteration of the mmRNA . target tissue . The composition contains an effective amount The level or concentration may be determined by a method of a ribonucleic acid engineered to avoid an innate immune such as , but not limited to , an assay using construct specific response of a cell into which the ribonucleic acid enters, probes , cytometry , qRT- PCR , realtime PCR , PCR , flow where the ribonucleic acid contains a nucleotide sequence cytometry, electrophoresis, mass spectrometry, or combina encoding a polypeptide of interest, under conditions such 50 tions thereof. Further, the level or concentration may be that the polypeptide of interest is produced in at least one associated with a clinical phenotype. For analysis , the exo target cell . The compositions generally contain a cell pen- some may be isolated by a method such as , but not limited etration agent, although “naked ” nucleic acid ( such as to , immunohistochemcial methods such as enzyme linked nucleic acids without a cell penetration agent or other agent) immunosorbant assay ( ELISA) methods, size exclusion is also contemplated , and a pharmaceutically acceptable 55 chromatography, density gradient centrifugation, differential carrier. centrifugation, nanomembrane ultrafiltration, immunoabsor In some circumstances, the amount of a protein produced bent capture, affinity purification, microfluidic separation, or by cells in a tissue is desirably increased . Preferably , this combinations thereof. increase in protein production is spatially restricted to cells Pharmaceutical Compositions within the target tissue . Thus, provided are methods of 60 When administered to a subject the pharmaceutical com increasing production of a protein of interest in a tissue of a positions described herein may provide proteins which have mammalian subject. A composition is provided that contains been generated from modified mRNAs. Pharmaceutical a ribonucleic acid that is engineered to avoid an innate compositions may optionally comprise one or more addi immune response of a cell into which the ribonucleic acid tional therapeutically active substances. In accordance with enters and encodes the polypeptide of interest and the 65 some embodiments, a method of administering pharmaceu composition is characterized in that a unit quantity of tical compositions comprising one or more proteins to be composition has been determined to produce the polypeptide delivered to a subject in need thereof is provided. In some US 10,898,574 B2 29 30 embodiments , compositions are administered to human sub- such as by producing any undesirable biological effect or jects . In a further embodiment, the compositions are administered to a subject who is a patient. Pharmaceutical compositions may optionally comprise one or more additional therapeutically active substances . In some embodiments, compositions are administered to humans. For the purposes of the present disclosure, the phrase “ active ingredient generally refers to a mmRNA to otherwise interacting in a deleterious manner with any other component ( s) of the pharmaceutical composition, its use is contemplated to be within the scope of this invention . 5 In some embodiments , a pharmaceutically acceptable excipient is at least 95 % , at least 96 % , at least 97 % , at least 98 % , at least 99 % , or 100 % pure . In some embodiments, an excipient is approved for use in humans and for veterinary be delivered as described herein . use . Although the descriptions of pharmaceutical composi- 10 tions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical com- 15 positions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood , and the ordinarily skilled veterinary pharmacologist can design and / or perform such modification with merely ordinary, if any, experimentation. 20 Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to , humans and / or other primates ; mammals, including commercially relevant mammals such as cattle, pigs , horses , sheep, cats, dogs , mice , and / or rats ; and / or birds , including 25 commercially relevant birds such as chickens, ducks, geese , and / or turkeys. Formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, 30 such preparatory methods include the step of bringing the active ingredient into association with an excipient and /or if nec one or more other accessory ingredients , and essary and / or desirable , shaping and / or packaging the prod35 uct into a desired single- or multi - dose unit. A pharmaceutical composition in accordance with the invention may be prepared, packaged , and / or sold in bulk , as a single unit dose , and / or as a plurality of single unit doses . As used herein, a " unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined 40 amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and / or a convenient fraction of such a dosage such as , for example, one -half or one - third of such a dosage . 45 Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and /or any additional ingredients in a pharmaceutical composition in accordance with the invention will vary, depending upon the identity, size , and / or condition of the subject treated and further depending 50 upon the route by which the composition is to be administered. By way of example, the composition may comprise between 0.1 % and 100% ( w / w ) active ingredient. Pharmaceutical formulations may additionally comprise a pharmaceutically acceptable excipient, which, as used 55 herein , includes any and all solvents, dispersion media , diluents, or other liquid vehicles, dispersion or suspension aids , surface active agents, isotonic agents, thickening or In some embodiments , an excipient is approved by United States Food and Drug Administration. In some embodi ments , an excipient is pharmaceutical grade. In some embodiments, an excipient meets the standards of the United States Pharmacopoeia ( USP ) , the European Pharmacopoeia (EP ) , the British Pharmacopoeia, and / or the International Pharmacopoeia . Pharmaceutically acceptable excipients used in the manu facture of pharmaceutical compositions include , but are not limited to , inert diluents, dispersing and /or granulating agents, surface active agents and /or emulsifiers, disintegrat ing agents , binding agents, preservatives, buffering agents , lubricating agents, and / or oils . Such excipients may option ally be included in pharmaceutical formulations. Excipients such as cocoa butter and suppository waxes , coloring agents , coating agents, sweetening, flavoring, and / or perfuming agents can be present in the composition, according to the judgment of the formulator. Exemplary diluents include, but are not limited to , cal cium carbonate, sodium carbonate, calcium phosphate , dicalcium phosphate , calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose , sucrose , cellulose , microcrystalline cellulose , kaolin, mannitol, sorbitol, inosi tol , sodium chloride, dry starch , cornstarch , powdered sugar, etc. , and / or combinations thereof. Exemplary granulating and / or dispersing agents include , but are not limited to , potato starch , corn starch , tapioca starch, sodium starch glycolate , clays , alginic acid , guar gum , citrus pulp , agar, bentonite, cellulose and wood prod ucts , natural sponge , cation -exchange resins , calcium car bonate, silicates, sodium carbonate, cross - linked poly ( vinyl pyrrolidone ) ( crospovidone), sodium carboxymethyl starch ( sodium starch glycolate ) , carboxymethyl cellulose , cross linked sodium carboxymethyl cellulose ( croscarmellose ), methylcellulose, pregelatinized starch ( starch 1500 ), micro crystalline starch , water insoluble starch , calcium car boxymethyl cellulose , magnesium aluminum silicate ( Veegum ), sodium lauryl sulfate , quaternary ammonium compounds, etc. , and / or combinations thereof. Exemplary surface active agents and / or emulsifiers include, but are not limited to , natural emulsifiers (e.g. acacia , agar, alginic acid, sodium alginate, tragacanth , chon drux , cholesterol , xanthan , pectin, gelatin, egg yolk, casein , wool fat, cholesterol, wax , and lecithin ), colloidal clays (e.g. bentonite [ aluminum silicate ) and Veegum [magnesium aluminum silicate ] ) , long chain amino acid derivatives, high molecular weight alcohols ( e.g. stearyl alcohol , cetyl alco hol , oleyl alcohol , triacetin monostearate, ethylene glycol distearate, glyceryl monostearate , and propylene glycol emulsifying agents, preservatives, solid binders, lubricants monostearate, polyvinyl alcohol ) , carbomers (e.g. carboxy and the like, as suited to the particular dosage form desired . 60 polymethylene, polyacrylic acid , acrylic acid polymer , and Remington's The Science and Practice of Pharmacy, 21st carboxyvinyl polymer ), carrageenan , cellulosic derivatives Edition, A. R. Gennaro ( Lippincott, Williams & Wilkins, (e.g. carboxymethylcellulose sodium , powdered cellulose , Baltimore, Md . , 2006 ; incorporated herein by reference ) hydroxymethyl cellulose , hydroxypropyl cellulose , discloses various excipients used in formulating pharmaceu- hydroxypropyl methylcellulose , methylcellulose ), sorbitan tical compositions and known techniques for the preparation 65 fatty acid esters ( e.g. polyoxyethylene sorbitan monolaurate thereof. Except insofar as any conventional excipient [ TWEEN?20 ), polyoxyethylene sorbitan [ TWEEN?60 ], medium is incompatible with a substance or its derivatives , polyoxyethylene sorbitan monooleate [ TWEEN®80] , sorbi US 10,898,574 B2 31 32 tan monopalmitate [ SPANR40 ), sorbitan monostearate PLUS® , PHENONIP® , methylparaben, GERMALL®115 , [ SPAN®60 ), sorbitan tristearate [SPAN®65 ], glyceryl GERMABEN?II, NEOLONETM , KATHONTM , and /or monooleate , sorbitan monooleate [SPAN®80 ]), polyoxyeth- EUXYL® . ylene esters (e.g. polyoxyethylene monostearate Exemplary buffering agents include , but are not limited [MYRJ®45 ], polyoxyethylene hydrogenated castor oil , 5 to , citrate buffer solutions , acetate buffer solutions, phos polyethoxylated castor oil , polyoxymethylene stearate, and buffer solutions , ammonium chloride, calcium carbon SOLUTOL® ), sucrose fatty acid esters , polyethylene glycol phate ate , calcium chloride , calcium citrate , calcium glubionate, fatty acid esters ( e.g. CREMOPHOR® ), polyoxyethylene calcium gluceptate , calcium gluconate, D - gluconic acid , ethers , ( e.g. polyoxyethylene lauryl ether [ BRIJR30 ] ) , poly calcium glycerophosphate , calcium lactate , propanoic acid , (nolamine vinyl-pyrrolidone ), diethylene glycol monolaurate, trietha. 10 calcium levulinate, pentanoic acid, dibasic calcium phos oleate , sodium oleate , potassium oleate , ethyl , phosphoric acid , tribasic calcium phosphate, calcium oleate, oleic acid, ethyl laurate, sodium lauryl sulfate , phate hydroxide phosphate, potassium acetate, potassium chlo PLURONIC®F 68 , POLOXAMER®188 , cetrimonium bro ride , potassium gluconate, potassium mixtures, dibasic mide, cetylpyridinium chloride , benzalkonium chloride, potassium phosphate , monobasic potassium phosphate , 15 docusate sodium , etc. and /or combinations thereof. Exemplary binding agents include, but are not limited to , potassium phosphate mixtures , sodium acetate, sodium starch ( e.g. cornstarch and starch paste) ; gelatin ; sugars (e.g. bicarbonate , sodium chloride, sodium citrate , sodium lac sucrose , glucose , dextrose, dextrin , molasses , lactose , lacti- tate , dibasic sodium phosphate , monobasic sodium phos tol , mannitol ); natural and synthetic gums (e.g. acacia , phate, sodium phosphate mixtures , tromethamine, magne sodium alginate, extract of Irish moss , panwar gum , ghatti 20 sium hydroxide, aluminum hydroxide, alginic acid , gum , mucilage of isapol husks, carboxymethylcellulose , pyrogen - free water, isotonic saline , Ringer's solution , ethyl methylcellulose, ethylcellulose, hydroxyethylcellulose, alcohol, etc. , and /or combinations thereof. hydroxypropyl cellulose , hydroxypropyl methylcellulose , Exemplary lubricating agents include, but are not limited microcrystalline cellulose , cellulose acetate, poly ( vinyl - pyr- to , magnesium stearate, calcium stearate , stearic acid, silica , rolidone ) , magnesium aluminum silicate ( VEEGUM® ), and 25 talc , malt , glyceryl behanate, hydrogenated vegetable oils , larch arabogalactan ); alginates; polyethylene oxide; poly- polyethylene glycol , sodium benzoate , sodium acetate , ethylene glycol ; inorganic calcium salts ; silicic acid ; polymethacrylates; waxes; water; alcohol, etc .; and combinations thereof. Exemplary preservatives may include , but are not limited to , antioxidants, chelating agents , antimicrobial preservatives , antifungal preservatives, alcohol preservatives, acidic preservatives, and / or other preservatives. Exemplary antioxidants include, but are not limited to , alpha tocopherol, ascorbic acid, acorbyl palmitate , butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite , propionic acid , propyl gallate , sodium ascorbate, sodium bisulfite, sodium metabisulfite , and /or sodium sulfite . Exemplary chelating agents include ethylenediaminetetraacetic acid ( EDTA ), citric acid monohydrate, disodium edetate , dipotassium edetate, edetic acid , fumaric acid , malic acid , phosphoric acid, sodium edetate, tartaric acid , and / or trisodium edetate. Exemplary antimicrobial preservatives include, but are not limited to , benzalkonium chloride , benzethonium chloride, benzyl alcohol , bronopol, cetrimide , cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol , ethyl alcohol , glycerin, hexetidine, imidurea , phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol , and / or thimerosal. Exemplary antifungal preservatives include , but are not limited to , butyl paraben , methyl paraben , ethyl paraben , propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate , potassium sorbate, sodium benzoate, sodium propionate, and / or sorbic acid . Exemplary alcohol preservatives include, but are not limited to , ethanol, polyethylene glycol, phenol, phenolic compounds , bisphenol, chlorobutanol, hydroxybenzoate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc. , and combinations thereof. Exemplary oils include , but are not limited to , almond , 30 apricot kernel, avocado , babassu, bergamot, black current seed, borage, cade , camomile , canola, caraway, carnauba , castor, cinnamon , cocoa butter, coconut, cod liver, coffee , corn , cotton seed, emu, eucalyptus, evening primrose , fish , flaxseed , geraniol, gourd , grape seed , hazel nut, hyssop , 35 isopropyl myristate, jojoba, kukui nut, lavandin , lavender, lemon, litsea cubeba, macademia nut, mallow , mango seed , meadowfoam seed , mink , nutmeg, olive, orange , orange roughy, palm , palm kernel, peach kernel, peanut, poppy seed, pumpkin seed , rapeseed, rice bran , rosemary, saf 40 flower, sandalwood, sasquana, savoury, sea buckthorn , sesame, shea butter, silicone , soybean , sunflower, tea tree , thistle, tsubaki , vetiver, walnut, and wheat germ oils . Exem plary oils include, but are not limited to , butyl stearate , caprylic triglyceride, capric triglyceride, cyclomethicone, 45 diethyl sebacate , dimethicone 360 , isopropyl myristate, min eral oil , octyldodecanol, oleyl alcohol , silicone oil , and / or combinations thereof. Liquid dosage forms for oral and parenteral administra tion include , but are not limited to , pharmaceutically accept 50 able emulsions, microemulsions, solutions, suspensions, syrups, and / or elixirs . In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art such as , for example, water or other solvents , solubilizing agents and emulsifiers such as ethyl alcohol , 55 isopropyl alcohol, ethyl carbonate, ethyl acetate , benzyl alcohol, benzyl benzoate , propylene glycol, 1,3 -butylene glycol , dimethylformamide, oils (in particular, cottonseed , and /or phenylethyl alcohol. Exemplary acidic preservatives groundnut, corn , germ , olive , castor, and sesame oils ) , include , but are not limited to , vitamin A , vitamin C , vitamin glycerol, tetrahydrofurfuryl alcohol , polyethylene glycols E , beta -carotene, citric acid , acetic acid , dehydroacetic acid , 60 and fatty acid esters of sorbitan, and mixtures thereof. ascorbic acid, sorbic acid , and /or phytic acid . Other preser- Besides inert diluents, oral compositions can include adju vatives include , but are not limited to , tocopherol, tocoph- vants such as wetting agents, emulsifying and suspending erol acetate , deteroxime mesylate , cetrimide, butylated agents, sweetening, flavoring, and /or perfuming agents . In hydroxyanisol ( BHA ) , butylated hydroxytoluened ( BHT ) , certain embodiments for parenteral administration , compo ethylenediamine, sodium lauryl sulfate ( SLS ) , sodium lauryl 65 sitions are mixed with solubilizing agents such as Cremo ether sulfate ( SLES ) , sodium bisulfite, sodium metabisulfite , phor® , alcohols , oils , modified oils , glycols , polysorbates, potassium sulfite, potassium metabisulfite , GLYDANT cyclodextrins, polymers , and / or combinations thereof. US 10,898,574 B2 33 General considerations in the formulation and / or manu- facture of pharmaceutical agents may be found, for example, in Remington : The Science and Practice of Pharmacy 21st ed . , Lippincott Williams & Wilkins, 2005 ( incorporated herein by reference ). In order to prolong the effect of an active ingredient, it is often desirable to slow the absorption of the active ingredient from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which , in turn, may depend upon crystal size and crystalline form . Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle . Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide - polyglycolide . Depending upon the ratio of drug to polymer and the nature of the particular polymer employed , the rate of drug release can be controlled . Examples of other biodegradable polymers include poly ( orthoesters ) and poly (anhydrides ). Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues . Compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing compositions with suitable non - irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient. Solid dosage forms for oral administration include capsules , tablets , pills , powders, and granules. In such solid dosage forms, an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/ or fillers or extenders ( e.g. starches, lactose , sucrose , glucose , mannitol, and silicic acid ), binders (e.g. carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone , sucrose , and acacia) , humectants (e.g. glycerol), disintegrating agents (e.g. agar, calcium carbonate, potato or tapioca starch , alginic acid, certain silicates, and sodium carbonate ), solution retarding agents ( e.g. paraffin ), absorption accelerators (e.g. quaternary ammonium compounds ), wetting agents (e.g. cetyl alcohol and glycerol monostearate ) , absorbents ( e.g. kaolin and bentonite clay ) , and lubricants ( e.g. talc , calcium stearate , magnesium stearate , solid polyethylene glycols , sodium lauryl sulfate ), and mixtures thereof. In the case of capsules , tablets and pills , the dosage form may comprise buffering agents . Solid compositions of a similar type may be employed as fillers in soft and hard - filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like . Solid dosage forms of tablets, dragees , capsules , pills , and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient ( s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner . Examples of embedding compositions which can be used include polymeric substances and waxes . Solid compositions of a similar type may be employed as fillers in soft and hard - filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. 34 Dosage forms for topical and /or transdermal administra 5 10 15 20 25 30 35 40 45 50 55 60 65 tion of a composition may include ointments , pastes , creams, lotions , gels , powders , solutions , sprays, inhalants and /or patches. Generally, an active ingredient is admixed under sterile conditions with a pharmaceutically acceptable excipient and / or any needed preservatives and / or buffers as may be required. Topically -administrable formulations may, for example, comprise from about 1 % to about 10 % ( w / w ) active ingredient, although the concentration of active ingre dient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical adminis tration may further comprise one or more of the additional ingredients described herein . A pharmaceutical composition may be prepared , pack aged , and / or sold in a formulation suitable for pulmonary administration via the buccal cavity. Such a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm . Such compositions are suitably in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant may be directed to disperse the powder and / or using a self propelling solvent/ powder dispensing container such as a device comprising the active ingredient dissolved and / or suspended in a low boiling propellant in a sealed container. Such powders comprise particles wherein at least 98 % of the particles by weight have a diameter greater than 0.5 nm and at least 95 % of the particles by number have a diameter less than 7 nm . Alternatively, at least 95 % of the particles by weight have a diameter greater than 1 nm and at least 90 % of the particles by number have a diameter less than 6 nm . Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form . Low boiling propellants generally include liquid propel lants having a boiling point of below 65 ° F. at atmospheric pressure . Generally the propellant may constitute 50% to 99.9 % ( w / w ) of the composition, and active ingredient may constitute 0.1 % to 20 % ( w / w ) of the composition. A pro pellant may further comprise additional ingredients such as a liquid non - ionic and / or solid anionic surfactant and / or a solid diluent ( which may have a particle size of the same order as particles comprising the active ingredient ). Pharmaceutical compositions formulated for pulmonary delivery may provide an active ingredient in the form of droplets of a solution and / or suspension . Such formulations may be prepared, packaged , and / or sold as aqueous and /or dilute alcoholic solutions and / or suspensions, optionally sterile, comprising active ingredient, and may conveniently be administered using any nebulization and / or atomization device . Such formulations may further comprise one or more additional ingredients including , but not limited to , a flavoring agent such as saccharin sodium , a volatile oil , a buffering agent, a surface active agent, and / or a preservative such as methylhydroxybenzoate. Droplets provided by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm . Formulations described herein as being useful for pulmo nary delivery are useful for intranasal delivery of a phar maceutical composition . Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 um to 500 um . Such a formulation is administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose . US 10,898,574 B2 36 35 Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1 % ( w / w ) and as much as 100 % ( w / w ) of active ingredient, and may comprise one or more of the additional ingredients described herein . A pharmaceutical composition may be prepared , packaged, 5 and / or sold in a formulation suitable for buccal administration . Such formulations may , for example , be in the form of tablets and / or lozenges made using conventional methods, and may, for example, 0.1 % to 20% ( w / w ) active ingredient, the balance comprising an orally dissolvable and / or degradable composition and, optionally, one or more of the additional ingredients described herein . Alternately, formulations suitable for buccal administration may comprise a powder and / or an aerosolized and / or atomized solution and / or suspension comprising active ingredient. Such powdered, aerosolized, and / or aerosolized formulations, when dispersed, may have an average particle and / or droplet size in the range from about 0.1 nm to about 200 nm , and may further comprise one or more of any additional ingredients described herein Properties of the Pharmaceutical Compositions The pharmaceutical compositions described herein can be characterized by one or more of the following properties : Bioavailability The mmRNA molecules , when formulated into a composition with a delivery agent as described herein , can exhibit an increase in bioavailability as compared to a composition lacking a delivery agent as described herein . As used herein , the term “ bioavailability ” refers to the systemic availability of a given amount of a mmRNA molecule administered to a mammal. Bioavailability can be assessed by measuring the area under the curve ( AUC ) or the maximum serum or plasma concentration (Cmar) of the unchanged form of a compound following administration of the compound to a mammal. AUC is a determination of the area under the curve plotting the serum or plasma concentration of a compound along the ordinate ( Y -axis) against time along the abscissa (X - axis ) . Generally, the AUC for a particular compound can be calculated using methods known to those of ordinary skill in the art and as described in G. S. Banker, Modern Pharmaceutics , Drugs and the Pharmaceutical Sciences , v. 72 , Marcel Dekker, New York , Inc. , 1996 , herein incorporated by reference. The Cmax value is the maximum concentration of the compound achieved in the serum or plasma of a mammal following administration of the compound to the mammal. The Cmax value of a particular compound can be measured using methods known to those of ordinary skill in the art. 10 15 20 25 30 35 40 45 composition as compared to the therapeutic window of the administered mmRNA molecule composition lacking a delivery agent as described herein . As used herein " thera peutic window ” refers to the range of plasma concentrations, or the range of levels of therapeutically active substance at the site of action , with a high probability of eliciting a therapeutic effect. In some embodiments, the therapeutic window of the mmRNA molecule when co - administered with a delivery agent as described herein can increase by at least about 2 % , at least about 5 % , at least about 10 % , at least about 15 % , at least about 20% , at least about 25 % , at least about 30 % , at least about 35 % , at least about 40 % , at least about 45 % , at least about 50% , at least about 55 % , at least about 60 % , at least about 65 % , at least about 70 % , at least about 75 % , at least about 80% , at least about 85 % , at least about 90 % , at least about 95 % , or about 100 % . Volume of Distribution The mmRNA molecules , when formulated into a compo sition as described herein , can exhibit an improved volume of distribution (V dist ). The volume of distribution (V dist) relates the amount of the drug in the body to the concen tration of the drug in the blood or plasma . As used herein , the term “ volume of distribution ” refers to the fluid volume that would be required to contain the total amount of the drug in the body at the same concentration as in the blood or plasma : Vdist equals the amount of drug in the body /concentration of drug in blood or plasma . For example, for a 10 mg dose and a plasma concentration of 10 mg / L , the volume of distribu tion would be 1 liter. The volume of distribution reflects the extent to which the drug is present in the extravascular tissue . A large volume of distribution reflects the tendency of a compound to bind to the tissue components compared with plasma protein binding . In a clinical setting, Vadist can be used to determine a loading dose to achieve a steady state concentration . In some embodiments , the volume of distri bution of the mmRNA molecule when co -administered with a delivery agent as described herein can decrease at least about 2 % , at least about 5 % , at least about 10 % , at least about 15 % , at least about 20% , at least about 25 % , at least about 30 % , at least about 35 % , at least about 40 % , at least about 45 % , at least about 50% , at least about 55 % , at least about 60 % , at least about 65 % , at least about 70 % . Devices and Methods for Multi - Administration Methods and devices for multi -administration may be employed to deliver the mmRNA of the present invention according to the split dosing regimens taught herein . Such methods and devices are described below. Method and devices known in the art for multi-adminis The phrases " increasing bioavailability " or " improving the tration to cells , organs and tissues are contemplated for use pharmacokinetics , " as used herein mean that the systemic 50 in conjunction with the methods and compositions disclosed availability of a first mmRNA molecule , measured as AUC , herein as embodiments of the present invention. These Cmax , or Cmin in a mammal is greater, when co - administered include, for example, those methods and devices having with a delivery agent as described herein , than when such multiple needles , hybrid devices employing for example co - administration does not take place . In some embodi- lumens or catheters as well as devices utilizing heat, electric ments , the bioavailability of the mmRNA molecule can 55 current or radiation driven mechanisms. increase by at least about 2 % , at least about 5 % , at least According to the present invention , these multi -adminis about 10% , at least about 15 % , at least about 20 % , at least about 25 % , at least about 30 % , at least about 35 % , at least about 40% , at least about 45 % , at least about 50 % , at least tration devices may be utilized to deliver the split doses contemplated herein . Suitable devices for use in delivering intradermal phar about 70% , at least about 75 % , at least about 80 % , at least needle devices such as those described in U.S. Pat . Nos . about 55 % , at least about 60 % , at least about 65 % , at least 60 maceutical compositions described herein include short about 85 % , at least about 90 % , at least about 95 % , or about 4,886,499 ; 5,190,521 ; 5,328,483 ; 5,527,288 ; 4,270,537 ; 5,015,235 ; 5,141,496 ; and 5,417,662 . Intradermal composi Therapeutic Window tions may be administered by devices which limit the The mmRNA molecules , when formulated into a compo- 65 effective penetration length of a needle into the skin , such as sition as described herein , can exhibit an increase in the those described in PCT publication WO 99/34850 and therapeutic window of the administered mmRNA molecule functional equivalents thereof. Jet injection devices which 100% . US 10,898,574 B2 38 37 deliver liquid compositions to the dermis via a liquid jet injector and /or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable . Jet injection devices are described , for example, in U.S. Pat . Nos . 5,480,381 ; 5,599,302 ; 5,334,144 ; 5,993,412 ; 5 5,649,912 ; 5,569,189 ; 5,704,911 ; 5,383,851 ; 5,893,397 ; 5,466,220 ; 5,339,163 ; 5,312,335 ; 5,503,627 ; 5,064,413 ; 5,520,639 ; 4,596,556 ; 4,790,824 ; 4,941,880 ; 4,940,460 ; and PCT publications WO 97/37705 and WO 97/13537 . Ballistic powder / particle delivery devices which use compressed gas to accelerate vaccine in powder form through the outer layers of the skin to the dermis are suitable . Alternatively or additionally, conventional syringes may be used in the classical mantoux method of intradermal administration . A method for delivering therapeutic agents to a solid tissue has been described by Bahrami et al and is taught for example in US Patent Publication 20110230839 , the contents of which are incorporated herein by reference in their entirety. According to Bahrami, an array of needles is incorporated into a device which delivers a substantially equal amount of fluid at any location in said solid tissue along each needle's length . A device for delivery of biological material across the biological tissue has been described by Kodgule et al and is taught for example in US Patent Publication 20110172610 , the contents of which are incorporated herein by reference in their entirety. According to Kodgule, multiple hollow microneedles made of one or more metals and having outer diameters from about 200 microns to about 350 microns and lengths of at least 100 microns are incorporated into the device which delivers peptides , proteins , carbohydrates, nucleic acid molecules , lipids and other pharmaceutically active ingredients or combinations thereof. A delivery probe for delivering a therapeutic agent to a tissue has been described by Gunday et al and is taught for example in US Patent Publication 20110270184 , the contents of which are incorporated herein by reference in their entirety. According to Gunday, multiple needles are incorporated into the device which moves the attached capsules between an activated position and an inactivated position to force the agent out of the capsules through the needles . A multiple -injection medical apparatus has been described by Assaf and is taught for example in US Patent Publication 20110218497 , the contents of which are incorporated herein by reference in their entirety. According to Assaf, multiple needles are incorporated into the device which has a chamber connected to one or more of said needles and a means for continuously refilling the chamber with the medical fluid after each injection . An at least partially implantable system for injecting a substance into a patient's body, in particular a penis erection stimulation system has been described by Forsell and is taught for example in US Patent Publication 20110196198 , the contents of which are incorporated herein by reference in their entirety . According to Forsell, multiple needles are incorporated into the device which is implanted along with one or more housings adjacent the patient's left and right corpora cavernosa . A reservoir and a pump are also implanted to supply drugs through the needles. A method for the transdermal delivery of a therapeutic effective amount of iron has been described by Berenson and is taught for example in US Patent Publication 20100130910 , the contents of which are incorporated herein by reference in their entirety. According to Berenson, multiple needles may be used to create multiple micro channels in stratum corneum to enhance transdermal delivery of the ionic iron on an iontophoretic patch . A method for delivery of biological material across the biological tissue has been described by Kodgule et al and is taught for example in US Patent Publication 20110196308 , the contents of which are incorporated herein by reference in their entirety. According to Kodgule , multiple biodegradable microneedles containing a therapeutic active ingredient are incorporated in a device which delivers proteins, carbohy drates, nucleic acid molecules , lipids and other pharmaceu tically active ingredients or combinations thereof. 10 A transdermal patch comprising a botulinum toxin com position has been described by Donovan and is taught for example in US Patent Publication 20080220020 , the con tents of which are incorporated herein by reference in their entirety. According to Donovan , multiple needles are incor 15 porated into the patch which delivers botulinum toxin under stratum corneum through said needles which project through the stratum corneum of the skin without rupturing a blood vessel . A cryoprobe for administration of an active agent to a 20 location of cryogenic treatment has been described by Toubia and is taught for example in US Patent Publication 20080140061 , the contents ofwhich are incorporated herein by reference in their entirety . According to Toubia , multiple needles are incorporated into the probe which receives the 25 active agent into a chamber and administers the agent to the tissue . A method for treating or preventing inflammation or promoting healthy joints has been described by Stock et al and is taught for example in US Patent Publication 30 20090155186 , the contents of which are incorporated herein by reference in their entirety. According to Stock , multiple needles are incorporated in a device which administers compositions containing signal transduction modulator compounds. 35 A multi - site injection system has been described by Kimmell et al and is taught for example in US Patent Publication 20100256594 , the contents of which are incor porated herein by reference in their entirety. According to Kimmell, multiple needles are incorporated into a device 40 which delivers a medication into a stratum corneum through the needles . A method for delivering interferons to the intradermal compartment has been described by Dekker et al and is taught for example in US Patent Publication 20050181033 , 45 the contents of which are incorporated herein by reference in 50 55 60 65 their entirety. According to Dekker, multiple needles having an outlet with an exposed height between 0 and 1 mm are incorporated into a device which improves pharmacokinet ics and bioavailability by delivering the substance at a depth between 0.3 mm and 2 mm . A method for delivering genes , enzymes and biological agents to tissue cells has described by Desai and is taught for example in US Patent Publication 20030073908 , the con tents of which are incorporated herein by reference in their entirety. According to Desai, multiple needles are incorpo rated into a device which is inserted into a body and delivers a medication fluid through said needles . A method for treating cardiac arrhythmias with fibroblast cells has been described by Lee et al and is taught for example in US Patent Publication 20040005295 , the con tents of which are incorporated herein by reference in their entirety. According to Lee , multiple needles are incorporated into the device which delivers fibroblast cells into the local region of the tissue . A method using a magnetically controlled pump for treating a brain tumor has been described by Shachar et al and is taught for example in U.S. Pat . No. 7,799,012 US 10,898,574 B2 39 40 (method ) and U.S. Pat . No. 7,799,016 (device ) , the contents uses a stretching assembly to enhance the contact of the of which are incorporated herein by reference in their needles with the skin and provides a more uniform delivery entirety. According Shachar, multiple needles were incorpo- of the substance . rated into the pump which pushes a medicating agent A perfusion device for localized drug delivery has been 5 described by Zamoyski and is taught for example in U.S. through the needles at a controlled rate . Methods of treating functional disorders of the bladder in Pat. No. 6,468,247 , the contents of which are incorporated mammalian females have been described by Versi et al and herein by reference in their entirety. According to Zamoyski, are taught for example in U.S. Pat . No. 8,029,496 , the multiple hypodermic needles are incorporated into the contents of which are incorporated herein by reference in device which injects the contents of the hypodermics into a their entirety. According to Versi, an array of micro - needles 10 tissue as said hypodermics are being retracted. is incorporated into a device which delivers a therapeutic A method for enhanced transport of drugs and biological agent through the needles directly into the trigone of the molecules across tissue by improving the interaction bladder. between micro -needles and human skin has been described A micro - needle transdermal transport device has been 15 by Prausnitz et al and is taught for example in U.S. Pat . No. described by Angel et al and is taught for example in U.S. 6,743,211 , the contents of which are incorporated herein by Pat . No. 7,364,568 , the contents of which are incorporated reference in their entirety. According to Prausnitz , multiple herein by reference in their entirety. According to Angel, micro -needles are incorporated into a device which is able to multiple needles are incorporated into the device which present a more rigid and less deformable surface to which transports a substance into a body surface through the 20 the micro - needles are applied. needles which are inserted into the surface from different directions . A device for intraorgan administration of medicinal agents has been described by Ting et al and is taught for A device for subcutaneous infusion has been described by example in U.S. Pat . No. 6,077,251 , the contents of which Dalton et al and is taught for example in U.S. Pat . No. are incorporated herein by reference in their entirety. 7,150,726 , the contents of which are incorporated herein by 25 According to Ting, multiple needles having side openings reference in their entirety. According to Dalton, multiple for enhanced administration are incorporated into a device needles are incorporated into the device which delivers fluid which by extending and retracting said needles from and into the needle chamber forces a medicinal agent from a through the needles into a subcutaneous tissue . reservoir into said needles and injects said medicinal agent A device and a method for intradermal delivery of vac 30 into a target organ . cines and gene therapeutic agents through microcannula A multiple needle holder and a subcutaneous multiple have been described by Mikszta et al and are taught for channel port has been described by Brown and is example in U.S. Pat . No. 7,473,247 , the contents of which taught forinfusion example in U.S. Pat . No. 4,695,273 , the contents are incorporated herein by reference in their entirety. of which are incorporated by reference in their According to Mitszta , at least one hollow micro - needle is 35 entirety . According to Brown, herein multiple on the needle incorporated into the device which delivers the vaccines to holder are inserted through the septumneedles of the infusion port the subject's skin to a depth ofbetween 0.025 mm and 2 mm . and communicate with isolated chambers in said infusion A method of delivering insulin has been described by port. Pettis et al and is taught for example in U.S. Pat. No. A dual hypodermic syringe has been described by Horn 7,722,595 , the contents of which are incorporated herein by 40 and is taught for example in U.S. Pat . No. 3,552,394 , the reference in their entirety . According to Pettis, two needles contents of which are incorporated herein by reference in are incorporated into a device wherein both needles insert their entirety. According to Horn , two needles incorporated essentially simultaneously into the skin with the first at a into the device are spaced apart less than 68 mm and may be depth of less than 2.5 mm to deliver insulin to intradermal of different styles and lengths, thus enabling injections to be compartment and the second at a depth of greater than 2.5 45 made to different depths. mm and less than 5.0 mm to deliver insulin to subcutaneous A syringe with multiple needles and multiple fluid com compartment. partments has been described by Hershberg and is taught for Cutaneous injection delivery under suction has been example in U.S. Pat . No. 3,572,336 , the contents of which described by Kochamba et al and is taught for example in are incorporated herein by reference in their entirety. U.S. Pat . No. 6,896,666 , the contents of which are incorpo- 50 According to Hershberg, multiple needles are incorporated rated herein by reference in their entirety. According to into the syringe which has multiple fluid compartments and Kochamba, multiple needles in relative adjacency with each is capable of simultaneously administering incompatible other are incorporated into a device which injects a fluid drugs which are not able to be mixed for one injection. below the cutaneous layer. A surgical instrument for intradermal injection of fluids A device for withdrawing or delivering a substance 55 has been described by Eliscu et al and is taught for example through the skin has been described by Down et al and is in U.S. Pat . No. 2,588,623 , the contents of which are taught for example in U.S. Pat . No. 6,607,513 , the contents incorporated herein by reference in their entirety. According of which are incorporated herein by reference in their to Eliscu , multiple needles are incorporated into the instru entirety. According to Down , multiple skin penetrating ment which injects fluids intradermally with a wider dis members which are incorporated into the device have 60 perse . lengths of about 100 microns to about 2000 microns and are An apparatus for simultaneous delivery of a substance to about 30 to 50 gauge. multiple breast milk ducts has been described by Hung and A device for delivering a substance to the skin has been is taught for example in EP 1818017 , the contents of which described by Palmer et al and is taught for example in U.S. are incorporated herein by reference in their entirety. Pat . No. 6,537,242 , the contents of which are incorporated 65 According to Hung, multiple lumens are incorporated into herein by reference in their entirety. According to Palmer, an the device which inserts though the orifices of the ductal array of micro -needles is incorporated into the device which networks and delivers a fluid to the ductal networks . US 10,898,574 B2 41 A catheter for introduction of medications to the tissue of a heart or other organs has been described by Tkebuchava and is taught for example in WO2006138109 , the contents of which are incorporated herein by reference in their entirety. According to Tkebuchava , two curved needles are incorporated which enter the organ wall in a flattened trajectory 42 A surgical device for ablating a channel and delivering at least one therapeutic agent into a desired region of the tissue has been described by McIntyre et al and is taught for example in U.S. Pat . No. 8,012,096 , the contents of which 5 are incorporated herein by reference in their entirety . According to McIntyre, multiple needles are incorporated into the device which dispenses a therapeutic agent into a Devices for delivering medical agents have been region of tissue surrounding the channel and is particularly described by Mckay et al and are taught for example in well suited for transmyocardial revascularization operations. WO2006118804 , the content of which are incorporated 10 Methods of treating functional disorders of the bladder in herein by reference in their entirety . According to Mckay, mammalian females have been described by Versi et al and multiple needles with multiple orifices on each needle are are taught for example in U.S. Pat . No. 8,029,496 , the incorporated into the devices to facilitate regional delivery contents of which are incorporated herein by reference in to a tissue , such as the interior disc space of a spinal disc . A method for directly delivering an immunomodulatory is their entirety. According to Versi, an array of micro -needles substance into an intradermal space within a mammalian is incorporated into a device which delivers a therapeutic skin has been described by Pettis and is taught for example agent through the needles directly into the trigone of the in WO2004020014 , the contents of which are incorporated bladder. herein by reference in their entirety. According to Pettis, A device and a method for delivering fluid into a flexible multiple needles are incorporated into a device which deliv- 20 biological barrier have been described by Yeshurun et al and ers the substance through the needles to a depth between 0.3 are taught for example in U.S. Pat . No. 7,998,119 (device) mm and 2 mm . and U.S. Pat . No. 8,007,466 (method ), the contents of which Methods and devices for administration of substances into are incorporated herein by reference in their entirety. at least two compartments in skin for systemic absorption According to Yeshurun , the micro -needles on the device and improved pharmacokinetics have been described by 25 penetrate and extend into the flexible biological barrier and Pettis et al and are taught for example in WO2003094995 , fluid is injected through the bore of the hollow micro the contents of which are incorporated herein by reference in needles . their entirety. According to Pettis , multiple needles having A method for epicardially injecting a substance into an lengths between about 300 um and about 5 mm are incor- area of tissue of a heart having an epicardial surface and porated into a device which delivers to intradermal and 30 disposed within a torso has been described by Bonner et al subcutaneous tissue compartments simultaneously. and is taught for example in U.S. Pat . No. 7,628,780 , the A drug delivery device with needles and a roller has been contents of which are incorporated herein by reference in described by Zimmerman et al and is taught for example in their entirety. According to Bonner, the devices have elon WO2012006259 , the contents of which are incorporated gate shafts and distal injection heads for driving needles into herein by reference in their entirety. According to Zimmer- 35 tissue and injecting medical agents into the tissue through man , multiple hollow needles positioned in a roller are the needles . incorporated into the device which delivers the content in a A device for sealing a puncture has been described by Nielsen et al and is taught for example in U.S. Pat . No. reservoir through the needles as the roller rotates . Methods and Devices Utilizing Catheters and/ or Lumens 7,972,358 , the contents of which are incorporated herein by Methods and devices using catheters and lumens may be 40 reference in their entirety. According to Nielsen, multiple employed to administer the mmRNA of the present inven- needles are incorporated into the device which delivers a tion on a split dosing schedule . Such methods and devices closure agent into the tissue surrounding the puncture tract. are described below. A method for myogenesis and angiogenesis has been A catheter -based delivery of skeletal myoblasts to the described by Chiu et al and is taught for example in U.S. Pat. myocardium of damaged hearts has been described by 45 No. 6,551,338 , the contents of which are incorporated herein Jacoby et al and is taught for example in US Patent Publi- by reference in their entirety. According to Chiu , 5 to 15 cation 20060263338 , the contents of which are incorporated needles having a maximum diameter of at least 1.25 mm and herein by reference in their entirety . According to Jacoby, a length effective to provide a puncture depth of 6 to 20 mm multiple needles are incorporated into the device at least part are incorporated into a device which inserts into proximity of which is inserted into a blood vessel and delivers the cell 50 with a myocardium and supplies an exogeneous angiogenic composition through the needles into the localized region of the subject's heart. An apparatus for treating asthma using neurotoxin has been described by Deem et al and is taught for example in or myogenic factor to said myocardium through the conduits which are in at least some of said needles. A method for the treatment of prostate tissue has been described by Bolmsj et al and is taught for example in U.S. US Patent Publication 20060225742 , the contents of which 55 Pat. No. 6,524,270 , the contents of which are incorporated are incorporated herein by reference in their entirety . herein by reference in their entirety. According to Bolmsj, a According to Deem , multiple needles are incorporated into device comprising a catheter which is inserted through the the device which delivers neurotoxin through the needles urethra has at least one hollow tip extendible into the into the bronchial tissue . surrounding prostate tissue . An astringent and analgesic A method for administering multiple -component thera- 60 medicine is administered through said tip into said prostate pies has been described by Nayak and is taught for example tissue . in U.S. Pat . No. 7,699,803 , the contents of which are A method for infusing fluids to an intraosseous site has incorporated herein by reference in their entirety. According been described by Findlay et al and is taught for example in to Nayak, multiple injection cannulas may be incorporated U.S. Pat. No. 6,761,726 , the contents of which are incorpo into a device wherein depth slots may be included for 65 rated herein by reference in their entirety. According to controlling the depth at which the therapeutic substance is Findlay, multiple needles are incorporated into a device delivered within the tissue . which is capable of penetrating a hard shell of material US 10,898,574 B2 44 43 covered by a layer of soft material and delivers a fluid at a electrodes to penetrate into the tissue surrounding said predetermined distance below said hard shell of material. A device for injecting medications into a vessel wall has lumen . Then the device introduces an agent through at least one of said needle electrodes and applies electric field by rated herein by reference in their entirety. According to Vigil, multiple injectors are mounted on each of the flexible tubes in the device which introduces a medication fluid through a multi - lumen catheter, into said flexible tubes and out of said injectors for infusion into the vessel wall . site . A delivery system for transdermal immunization has been described by Levin et al and is taught for example in been described by Vigil et al and is taught for example in said pair of needle electrodes to allow said agent pass U.S. Pat . No. 5,713,863 , the contents of which are incorpo- 5 through the cell membranes into the cells at the treatment 10 A catheter for delivering therapeutic and / or diagnostic agents to the tissue surrounding a bodily passageway has WO2006003659 , the contents of which are incorporated multiple electrodes are incorporated into the device which applies electrical energy between the electrodes to generate herein by reference in their entirety. According to Levin , been described by Faxon et al and is taught for example in micro channels in the skin to facilitate transdermal delivery. U.S. Pat . No. 5,464,395 , the contents of which are incorpo A method for delivering RF energy into skin has been rated herein by reference in their entirety. According to 15 described by Schomacker and is taught for example in Faxon , at least one needle cannula is incorporated into the WO2011163264 , the contents of which are incorporated catheter which delivers the desired agents to the tissue through said needles which project outboard of the catheter. herein by reference in their entirety. According to Schoma Balloon catheters for delivering therapeutic agents have cker, multiple needles are incorporated into a device which been described by Orr and are taught for example in 20 applies vacuum to draw skin into contact with a plate so that WO2010024871 , the contents of which are incorporated needles insert into skin through the holes on the plate and herein by reference in their entirety. According to Orr, deliver RF energy. multiple needles are incorporated into the devices which Devices and Kits deliver the therapeutic agents to different depths within the Devices may also be used in conjunction with the present tissue . 25 invention. In one embodiment, a device is used to assess levels of a protein which has been administered in the form Methods and Devices Utilizing Electrical Current Methods and devices utilizing electric current may be of a modified mRNA . The device may comprise a blood , employed to deliver the mmRNA of the present invention urine or other biofluidic test . It may be as large as to include according to the split dosing regimens taught herein . Such an automated central lab platform or a small decentralized methods and devices are described below. 30 bench top device . It may be point of care or a handheld An electro collagen induction therapy device has been device. The device may be useful in drug discovery efforts described by Marquez and is taught for example in US as a companion diagnostic. Patent Publication 20090137945 , the contents of which are incorporated herein by reference in their entirety. According to Marquez, multiple needles are incorporated into the device which repeatedly pierce the skin and draw in the skin a portion of the substance which is applied to the skin first. An electrokinetic system has been described by Etheredge et al and is taught for example in US Patent Publication 20070185432 , the contents ofwhich are incorporated herein by reference in their entirety. According to Etheredge, micro -needles are incorporated into a device which drives by an electrical current the medication through the needles into the targeted treatment site . An iontophoresis device has been described by Matsumura et al and is taught for example in U.S. Pat . No. 7,437,189 , the contents of which are incorporated herein by reference in their entirety. According to Matsumura, multiple needles are incorporated into the device which is capable of delivering ionizable drug into a living body at higher speed or with higher efficiency. Intradermal delivery of biologically active agents by needle - free injection and electroporation has been described by Hoffmann et al and is taught for example in U.S. Pat . No. 7,171,264 , the contents of which are incorporated herein by reference in their entirety. According to Hoffmann, one or more needle - free injectors are incorporated into an electroporation device and the combination of needle - free injec- In some embodiments the device is self -contained , and is 35 40 45 50 55 tion and electroporation is sufficient to introduce the agent into cells in skin , muscle or mucosa . 60 A method for electropermeabilization -mediated intracel- optionally capable of wireless remote access to obtain instructions for synthesis and / or analysis of the generated nucleic acid . The device is capable of mobile synthesis of at least one nucleic acid , and preferably an unlimited number of different nucleic acid sequences. In certain embodiments , the device is capable of being transported by one or a small number of individuals. In other embodiments, the device is scaled to fit on a benchtop or desk . In other embodiments , the device is scaled to fit into a suitcase , backpack or similarly sized object. In further embodiments, the device is scaled to fit into a vehicle, such as a car , truck or ambulance, or a military vehicle such as a tank or personnel carrier. The information necessary to generate a modified mRNA encod ing protein of interest is present within a computer readable medium present in the device . In some embodiments, the device is capable of commu nication ( e.g. , wireless communication ) with a database of nucleic acid and polypeptide sequences. The device contains at least one sample block for insertion of one or more sample vessels . Such sample vessels are capable of accepting in liquid or other form any number of materials such as template DNA , nucleotides , enzymes, buffers, and other reagents. The sample vessels are also capable of being heated and cooled by contact with the sample block . The sample block is generally in communication with a device base with one or more electronic control units for the at least one sample block . The sample block preferably contains a heating module, such heating molecule capable of heating and / or cooling the sample vessels and contents thereof to temperatures between about -20 C and above +100 C. The device base is in communication with a voltage supply such as a battery or external voltage supply. The device also contains means for storing and distributing the materials for lular delivery has been described by Lundkvist et al and is taught for example in U.S. Pat . No. 6,625,486 , the contents of which are incorporated herein by reference in their entirety. According to Lundkvist, a pair of needle electrodes 65 is incorporated into a catheter. Said catheter is positioned into a body lumen followed by extending said needle RNA synthesis. US 10,898,574 B2 46 45 Optionally, the sample block contains a module for separating the synthesized nucleic acids . Alternatively, the device contains a separation module operably linked to the sample block . Preferably the device contains a means for analysis of the synthesized nucleic acid . Such analysis 5 example , the term “ C1-6 alkyl ” is specifically intended to individually disclose methyl, ethyl, C , alkyl, C4 alkyl, Cg alkyl, and Co alkyl. Animal: As used herein , the term “ animal ” refers to any member of the animal kingdom . In some embodiments, includes sequence identity ( demonstrated such as by hybrid “ animal ” refers to humans at any stage of development. In ization) , absence of non -desired sequences, measurement of some embodiments, " animal” refers to non - human animals integrity of synthesized mRNA ( such has by microfluidic at any stage of development. In certain embodiments, the - human animal is a mammal (e.g. , a rodent, a mouse , a viscometry combined with spectrophotometry ), and concen 10 non , a rabbit, a monkey, a dog , a cat , a sheep, cattle, a primate , tration and /orpotency of modified RNA ( such as by spec rat or a pig ) . In some embodiments, animals include , but are not trophotometry ). limited to , mammals, birds , reptiles, amphibians, fish , and In certain embodiments, the device is combined with a worms In some embodiments, the animal is a transgenic means for detection of pathogens present in a biological animal,. genetically - engineered animal, or a clone . material obtained from a subject, e.g. , the IBIS PLEX - ID 15 Approximately : As used herein , the term “ approximately ” system ( Abbott) for microbial identification . or “ about, ” as applied to one or more values of interest, The present invention provides for devices which incor refers to a value that is similar to a stated reference value . In porate mmRNA that encode proteins of interest. These certain embodiments, the term “ approximately ” or “ about” devices may be implantable in an animal subject or may refers to a range of values that fall within 25 % , 20 % , 19 % , supply mmRNA formulations via a catheter or lumen . The 20 18 % , 17 % , 16 % , 15 % , 14 % , 13 % , 12 % , 11 % , 10 % , 9 % , 8 % , device may be connected to or incorporate a pump. Such 7 % , 6 % , 5 % , 4 % , 3 % , 2 % , 1 % , or less in either direction devices include those which can deliver therapeutics to areas (greater than or less than ) of the stated reference value unless of the body not readily accessible such the CNS or across otherwise stated or otherwise evident from the context the blood brain barrier. In this embodiment the split dosing (except where such number would exceed 100 % of a pos 25 sible value) . regimen can be implemented using a regulated pump. Associated with : As used herein , the terms " associated Kits ,” “ conjugated " " linked ," " attached , ” and “ tethered , " The invention provides a variety of kits for conveniently with and / or effectively carrying out methods of the present inven when used with respect to two or more moieties , means that moieties are physically associated or connected with one tion . Typically kits will comprise sufficient amounts and / or 30 the another, either directly or via one or more additional moi numbers of components to allow a user to perform multiple eties that serves as a linking agent, to form a structure that treatments of a subject( s ) and /or to perform multiple experi is sufficiently stable so that the moieties remain physically ments. associated under the conditions in which the ructure is In one aspect , the present invention provides kits for , e.g. , physiological conditions . An “ association ” need protein production , comprising a first isolated nucleic acid 35 used not be through direct covalent chemical bonding . It comprising a translatable region and a nucleic acid modifi may alsostrictly suggest ionic or hydrogen bonding or a hybrid cation , wherein the nucleic acid may be capable of evading ization based connectivity sufficiently stable such that the an innate immune response of a cell into which the first “ associated ” entities remain physically associated . isolated nucleic acid may be introduced , and packaging and Bifunctional: As used herein , the term “ bifunctional” instructions. The kit may further comprise a delivery agent 40 refers to any substance , molecule or moiety which is capable to form a formulation composition. The delivery composi- of or maintains at least two functions. The functions may tion may comprise a lipidoid. The lipoid may be selected from , but is not limited to , C12-200 , 98N12-5 , MD1 , DLinDMA , DLin - K -DMA, DLin -KC2 -DMA , DLin -MC3 - DMA 45 and analogs thereof. effect the same outcome or a different outcome. The struc ture that produces the function may be the same or different. For example , bifunctional modified RNAs of the present invention may encode a cytotoxic peptide (a first function ) while those nucleosides which comprise the encoding RNA are, in and of themselves, cytotoxic ( second function ). In this example , delivery of the bifunctional modified RNA to a cancer cell would produce not only a peptide or protein In one aspect , the present invention provides kits for protein production , comprising a first isolated nucleic acid comprising a translatable region and a nucleoside modification , wherein the nucleic acid exhibits reduced degradation by a cellular nuclease , and packaging and instructions. 50 molecule which may ameliorate or treat the cancer but In one aspect , the present invention provides kits for would also deliver a cytotoxic payload of nucleosides to the protein production , comprising a first isolated nucleic acid cell should degradation, instead of translation of the modi comprising a translatable region and at least two different fied RNA , occur. nucleoside modifications , wherein the nucleic acid exhibits Biologically active : As used herein , the phrase " biologi reduced degradation by a cellular nuclease, and packaging 55 cally active ” refers to a characteristic of any substance that has activity in a biological system and /or organism . For and instructions. In some embodiments, kits would provide split doses or instructions for the administration of split dosages of the mmRNA of the kit . instance, a substance that, when administered to an organ ism , has a biological affect on that organism , is considered to be biologically active . In particular embodiments, a 60 nucleic acid molecule of the present invention may be considered biologically active if even a portion of the nucleic acid molecule is biologically active or mimics an At various places in the present specification, substituents activity considered biologically relevant. of compounds of the present disclosure are disclosed in Chemical terms: As used herein, the term “ alkyl” is meant groups or in ranges. It is specifically intended that the 65 to refer to a saturated hydrocarbon group which is straight present disclosure include each and every individual sub- chained or branched . Example alkyl groups include methyl combination of the members of such groups and ranges . For (Me) , ethyl (Et) , propyl (e.g. , n -propyl and isopropyl ), butyl Definitions US 10,898,574 B2 47 48 ( e.g. , n -butyl, isobutyl, t -butyl ), pentyl ( e.g. , n -pentyl, isopentyl, neopentyl), and the like . An alkyl group can contain different number of neutrons in the nuclei. For example, isotopes of hydrogen include tritium and deuterium . from 1 to about 20 , from 2 to about 20 , from 1 to about 12 , from 1 to about 8 , from 1 to about 6 , from 1 to about 4 , or The compounds and salts of the present disclosure can be prepared in combination with solvent or water molecules to 5 form solvates and hydrates by routine methods . from 1 to about 3 carbon atoms. Conserved : As used herein , the term " conserved ” refers to As used herein , " alkenyl” refers to an alkyl group having or amino acid residues of a polynucleotide one or more double carbon - carbon bonds . Example alkenyl nucleotides sequence or polypeptide sequence, respectively, that are groups include ethenyl, propenyl, and the like . occur unaltered in the same position of two or As used herein , “ alkoxy ” refers to an O -alkyl group. those that sequences being compared. Nucleotides or amino Example alkoxy groups include methoxy, ethoxy, propoxy 10 more acids that are relatively conserved are those that are con ( e.g. , n - propoxy and isopropoxy ), t - butoxy, and the like . served amongst more related sequences than nucleotides or As used herein , “ alkenyl ” refers to an alkyl, as defined amino acids appearing elsewhere in the sequences . above , containing at least one double bond between adjacent In some embodiments, two or more sequences are said to carbon atoms. Alkenyls include both cis and trans isomers. 15 be “completely conserved” if they are 100% identical to one Representative straight chain and branched alkenyls include another. In some embodiments, two or more sequences are ethylenyl, propylenyl, 1 - butenyl, 2 -butenyl, isobutylenyl, 1 -pentenyl, 2 -pentenyl, 3 -methyl - 1 -butenyl, 2-methyl-2butenyl, 2,3 - dimethyl - 2 - butenyl , and the like . said to be “ highly conserved ” if they are at least 70 % identical, at least 80% identical, at least 90 % identical, or at least 95 % identical to one another. In some embodiments, As used herein, “ alkynyl” refers to an alkyl group having 20 two or more sequences are said to be “ highly conserved ” if one or more triple carbon - carbon bonds . Example alkynyl they are about 70 % identical, about 80% identical, about groups include ethynyl, propynyl, and the like . 90% identical, about 95 % , about 98 % , or about 99 % iden As used herein , " aryl” refers to monocyclic or polycyclic tical to one another. In some embodiments, two or more ( e.g. , having 2 , 3 or 4 fused rings ) aromatic hydrocarbons sequences are said to be “ conserved ” if they are at least 30 % such as , for example, phenyl, naphthyl, anthracenyl, 25 identical, at least 40 % identical, at least 50% identical, at phenanthrenyl, indanyl, indenyl, and the like. In some least 60% identical, at least 70 % identical, at least 80 % embodiments, aryl groups have from 6 to about 20 carbon identical, at least 90% identical, or at least 95 % identical to atoms . one another. In some embodiments , two or more sequences As used herein , “ halo ” or “ halogen ” includes fluoro , are said to be " conserved ” if they are about 30 % identical, chloro , bromo , and iodo . 30 about 40 % identical, about 50% identical, about 60 % iden Compound: As used herein , the term “ compound,” is tical , about 70% identical, about 80% identical, about 90 % meant to include all stereoisomers , geometric isomers, tau- identical, about 95 % identical, about 98 % identical, or about 99 % identical to one another. Conservation of sequence may tomers , and isotopes of the structures depicted . The compounds described herein can be asymmetric ( e.g. , apply to the entire length of an oligonucleotide or polypep having one or more stereocenters ). All stereoisomers, such 35 tide or may apply to a portion, region or feature thereof. as enantiomers and diastereomers , are intended unless otherwise indicated . Compounds of the present disclosure that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. Many geometric isomers of olefins, C=N double bonds, and the like can also be present in the compounds described herein , and all such stable isomers are contemplated in the present disclosure . Cis and trans geometric isomers of the compounds of the present disclosure are described and may be isolated as a mixture of isomers or as separated isomeric forms. Compounds of the present disclosure also include tautomeric forms. Tautomeric forms result from the swapping of a single bond with an adjacent double bond and the concomitant migration of a proton . Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge. Examples prototropic tautomers include ketone - enol pairs , amide - imidic acid pairs , lactam - lactim pairs , amide - imidic acid pairs , enamine - imine pairs , and annular forms where a proton can occupy two or more positions of a heterocyclic system , such as , 1H- and 3H - imidazole , 1H- , 2H- and 4H - 1,2,4 -triazole, 1H- and 2H - isoindole, and 1H- and 2H -pyrazole. Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution . Compounds of the present disclosure also include all of the isotopes of the atoms occurring in the intermediate or final compounds. “ Isotopes ” refers to atoms having the same atomic number but different mass numbers resulting from a 40 45 50 55 60 65 Delivery : As used herein , “ delivery ” refers to the act or manner of delivering a compound, substance, entity , moiety, cargo or payload . Delivery Agent: As used herein , “ delivery agent ” refers to any substance which facilitates, at least in part, the in vivo delivery of a nucleic acid molecule to targeted cells . Detectable label : As used herein , " detectable label” refers to one or more markers , signals, or moieties which are attached , incorporated or associated with another entity that is readily detected by methods known in the art including radiography , fluorescence, chemiluminescence, enzymatic activity, absorbance and the like . Detectable labels include radioisotopes, fluorophores, chromophores, enzymes , dyes , metal ions , ligands such as biotin, avidin , strepavidin and haptens, quantum dots , and the like . Detectable labels may be located at any position in the peptides or proteins dis closed herein . They may be within the amino acids , the peptides , or proteins, or located at the N- or C - termini. Distal : As used herein " distal ” means farther from center mass or line of symmetry of subject or reference point. For limbs, it is farther from body. Dosing regimen : As used herein , a “ dosing regimen ” is a schedule of administration or physician determined regimen of treatment, prophylaxis, or palliative care . Dose splitting factor (DSF )-ratio of PUD of dose split treatment divided by PUD of total daily dose or single unit dose . The value is derived from comparison of dosing regimens groups. Expression: As used herein , “ expression ” of a nucleic acid sequence refers to one or more of the following events : ( 1 ) production of an RNA template from a DNA sequence ( e.g. , by transcription ); (2 ) processing of an RNA transcript ( e.g. , US 10,898,574 B2 49 50 by splicing , editing , 5 ' cap formation , and / or 3 ' end processing ) ; ( 3 ) translation of an RNA into a polypeptide or protein ; and (4 ) post - translational modification of a polypeptide or account the number of gaps, and the length of each gap , which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determi protein . nation of percent identity between two sequences can be Formulation : As used herein , a “ formulation ” includes at 5 accomplished using a mathematical algorithm . For example, least a modified nucleic acid molecule and a delivery agent. the percent identity between two nucleotide sequences can Functional : As used herein , a “ functional ” biological be determined using methods such as those described in molecule is a biological molecule in a form in which it exhibits a property and / or activity by which it is characterized. Homology : As used herein , the term “ homology ” refers to the overall relatedness between polymeric molecules , e.g. between nucleic acid molecules ( e.g. DNA molecules and /or RNA molecules ) and / or between polypeptide molecules . In some embodiments, polymeric molecules are considered to be “ homologous” to one another if their sequences are at least 25 % , at least 30 % , at least 35 % , at least 40 % , at least 45 % , at least 50 % , at least 55 % , at least 60 % , at least 65 % , at least 70 % , at least 75 % , at least 80 % , at least 85 % , at least 90 % , at least 95 % , or at least 99 % identical. In some embodiments, polymeric molecules are considered to be “ homologous ” to one another if their sequences are at least 25 % , at least 30 % , at least 35 % , at least 40 % , at least 45 % , at least 50 % , at least 55 % , at least 60 % , at least 65 % , at least 70 % , at least 75 % , at least 80 % , at least 85 % , at least 90 % , at least 95 % , or at least 99 % similar. The term “ homologous” necessarily refers to a comparison between at least two sequences ( polynucleotide or polypeptide sequences ). In accordance with the invention, two polynucleotide sequences are considered to be homologous if the polypeptides they encode are at least about 50% identical, at least about 60 % identical, at least about 70 % identical, at least about 80 % identical, or at least about 90 % identical for least one stretch of at least about 20 amino acids . In some embodiments, homologous polynucleotide sequences are characterized by the ability to encode a stretch of at least 4-5 uniquely specified amino acids . For polynucleotide sequences less than 60 nucleotides in length , homology is determined by the ability to encode a stretch of at least 4-5 uniquely specified amino acids . In accordance with the invention , two protein sequences are considered to be homologous if the proteins are at least about 50 % identical, at least about 60 % identical, at least about 70 % identical, at least about 80 % identical, or at least about 90% identical for at least one stretch of at least about 20 amino acids . Identity: As used herein , the term “ identity ” refers to the overall relatedness between polymeric molecules , e.g. , between oligonucleotide molecules ( e.g. DNA molecules and / or RNA molecules ) and / or between polypeptide molecules . Calculation of the percent identity of two polynucleotide sequences, for example, can be performed by aligning the two sequences for optimal comparison purposes ( e.g. , gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non- identical sequences can be disregarded for comparison purposes ) . In certain embodiments, the length of a sequence aligned for comparison purposes is at least 30% , at least Computational Molecular Biology, Lesk , A. M. , ed . , Oxford University Press, New York, 1988 ; Biocomputing: Informat 10 ics and Genome Projects, Smith, D. W., ed . , Academic Press, New York , 1993 ; Sequence Analysis in Molecular Biology, von Heinje , G. , Academic Press , 1987 ; Computer Analysis of Sequence Data, Part I , Griffin , A. M. , and Griffin , H. G. , eds . , Humana Press, New Jersey, 1994 ; and Sequence 15 Analysis Primer, Gribskov, M. and Devereux , J. , eds . , M Stockton Press , New York , 1991 ; each of which is incorpo rated herein by reference. For example, the percent identity between two nucleotide sequences can be determined using the algorithm of Meyers and Miller (CABIOS , 1989 , 4 :11 20 17 ) , which has been incorporated into the ALIGN program ( version 2.0 ) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can , alterna tively, be determined using the GAP program in the GCG 25 software package using an NWSgapdna.CMP matrix . Meth ods commonly employed to determine percent identity between sequences include , but are not limited to those disclosed in Carillo , H. , and Lipman , D. , SIAM J Applied Math ., 48 : 1073 ( 1988 ) ; incorporated herein by reference . 30 Techniques for determining identity are codified in publicly available computer programs. Exemplary computer soft ware to determine homology between two sequences include, but are not limited to , GCG program package , Devereux, J. , et al . , Nucleic Acids Research , 12 ( 1 ) , 387 35 ( 1984 ) ) , BLASTP, BLASTN , and FASTA Atschul, S. F. et al . , J. Molec. Biol., 215 , 403 ( 1990 ) ) . Inhibit expression of a gene: As used herein , the phrase “ inhibit expression of a gene” means to cause a reduction in the amount of an expression product of the gene. The 40 expression product can be an RNA transcribed from the gene (e.g. , an mRNA ) or a polypeptide translated from an mRNA transcribed from the gene. Typically a reduction in the level of an mRNA results in a reduction in the level of a polypeptide translated therefrom . The level of expression 45 may be determined using standard techniques for measuring mRNA or protein . In vitro : As used herein , the term " in vitro ” refers to events that occur in an artificial environment, e.g. , in a test tube or reaction vessel , in cell culture, in a Petri dish, etc., 50 rather than within an organism ( e.g. , animal, plant, or microbe ) . In vivo : As used herein , the term " in vivo ” refers to events that occur within an organism (e.g. , animal, plant, or microbe or cell or tissue thereof). 55 Isolated : As used herein , the term “ isolated ” refers to a substance or entity that has been separated from at least some of the components with which it was associated ( whether in nature or in an experimental setting ). Isolated 40 % , at least 50 % , at least 60 % , at least 70 % , at least 80 % , substances may have varying levels of purity in reference to at least 90 % , at least 95 % , or 100 % of the length of the 60 the substances from which they have been associated . Iso reference sequence. The nucleotides at corresponding lated substances and / or entities may be separated from at nucleotide positions are then compared . When a position in least about 10 % , about 20 % , about 30% , about 40 % , about the first sequence is occupied by the same nucleotide as the 50% , about 60 % , about 70 % , about 80 % , about 90% , or corresponding position in the second sequence, then the more of the other components with which they were initially molecules are identical at that position . The percent identity 65 associated . In some embodiments, isolated agents are more between the two sequences is a function of the number of than about 80 % , about 85 % , about 90 % , about 91 % , about identical positions shared by the sequences , taking into 92 % , about 93 % , about 94 % , about 95 % , about 96 % , about US 10,898,574 B2 51 52 97 % , about 98 % , about 99 % , or more than about 99 % pure . Pharmaceutically acceptable salts : The present disclosure As used herein, a substance is “ pure ” if it is substantially free also includes pharmaceutically acceptable salts of the com of other components. Substantially isolated : By “ substan- pounds described herein . As used herein , “ pharmaceutically tially isolated” is meant that the compound is substantially acceptable salts ” refers to derivatives of the disclosed com separated from the environment in which it was formed or 5 pounds wherein the parent compound is modified by con detected . Partial separation can include, for example, a verting an existing acid or base moiety to its salt form . composition enriched in the compound of the present dis- Examples of pharmaceutically acceptable salts include, but closure . Substantial separation can include compositions are not limited to , mineral or organic acid salts of basic containing at least about 50 % , at least about 60 % , at least residues such as amines ; alkali or organic salts of acidic about 70% , at least about 80 % , at least about 90 % , at least 10 residues such as carboxylic acids ; and the like . The phar about 95 % , at least about 97 % , or at least about 99 % by maceutically acceptable salts of the present disclosure weight of the compound of the present disclosure, or salt include the conventional non - toxic salts of the parent com thereof. Methods for isolating compounds and their salts are pound formed , for example, from non - toxic inorganic or routine in the art. organic acids . The pharmaceutically acceptable salts of the Modified: As used herein “ modified ” refers to a changed 15 present disclosure can be synthesized from the parent com state or structure of a molecule of the invention . Molecules pound which contains a basic or acidic moiety by conven may be modified in many ways including chemically, struc- tional chemical methods. Generally, such salts can be pre turally, and functionally. In one embodiment, the mRNA pared by reacting the free acid or base forms of these molecules of the present invention are modified by the compounds with a stoichiometric amount of the appropriate introduction of non - natural nucleosides and / or nucleotides. 20 base or acid in water or in an organic solvent, or in a mixture Modified , as it pertains to a modified mRNA may also mean of the two; generally, nonaqueous media like ether, ethyl acetate , ethanol, isopropanol , or acetonitrile are preferred . any alteration which is different from the wild type. Naturally occurring: As used herein , “ naturally occurring " Lists of suitable salts are found in Remington's Pharmaceu means existing in nature without artificial aid . tical Sciences, 17th ed . , Mack Publishing Company, Easton , Patient: As used herein , “ patient ” refers to a subject who 25 Pa . , 1985 , p . 1418 and Journal of Pharmaceutical Science, may seek or be in need of treatment, requires treatment, is 66 , 2 ( 1977 ) , each of which is incorporated herein by receiving treatment, will receive treatment, or a subject who reference in its entirety . is under care by a trained professional for a particular Polypeptide: As used herein , “ polypeptide” means a poly disease or condition . mer of amino acid residues linked together by peptide bonds. Peptide : As used herein, “ peptide ” is less than or equal to 30 The term , as used herein, refers to proteins, polypeptides , 50 amino acids long , e.g. , about 5 , 10 , 15 , 20 , 25 , 30 , 35 , 40 , and peptides of any size , structure, or function . Typically, 45 , or 50 amino acids long . however, a polypeptide will be at least 50 amino acids long . Prodrug: The present disclosure also includes prodrugs of In some instances the polypeptide encoded is smaller than the compounds described herein . As used herein , “ prodrugs ” about 50 amino acids and the polypeptide is termed a refer to any substance , molecule or entity which is in a form 35 peptide. If the polypeptide is a peptide, it will be at least predicate for that substance, molecule or entity to act as a about 5 amino acid residues long. Thus, polypeptides therapeutic upon chemical or physical alteration . Prodrugs include gene products, naturally occurring polypeptides, may by covalently bonded or sequestested in some way and synthetic polypeptides, homologs , orthologs, paralogs, frag which release or are converted into the active drug moiety ments and other equivalents, variants, and analogs of the prior to , upon or after administered to a mammalian subject. 40 foregoing. A polypeptide may be a single molecule or may Prodrugs can be prepared by modifying functional groups be a multi-molecular complex such as a dimer, trimer or present in the compounds in such a way that the modifica- tetramer . The term polypeptide may also apply to amino acid tions are cleaved , either in routine manipulation or in vivo , polymers in which one or more amino acid residues are an to the parent compounds. Prodrugs include compounds artificial chemical analogue of a corresponding naturally wherein hydroxyl, amino , sulfhydryl, or carboxyl groups are 45 occurring amino acid . bonded to any group that, when administered to a mammaPolypeptide per unit drug (PUD ) : As used herein , a PUD lian subject, cleaves to form a free hydroxyl, amino , sulf- or product per unit drug, is defined as a subdivided portion hydryl, or carboxyl group respectively. Preparation and use of total daily dose , usually 1 mg , pg , kg , etc. , of a product of prodrugs is discussed in T. Higuchi and V. Stella , “ Pro- ( such as a polypeptide ) as measured in body fluid or tissue , drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. 50 usually defined in concentration such as pmol/mL , mmol / Symposium Series, and in Bioreversible Carriers in Drug mL , etc divided by the measure in the body fluid . Design, ed . Edward B. Roche , American Pharmaceutical Proximal : As used herein , “ proximal” means closer to Association and Pergamon Press , 1987 , both of which are center mass or line of symmetry of subject or reference hereby incorporated by reference in their entirety. point. For limbs, it is closer to body. Proliferate: As used herein , the term “ proliferate ” means 55 Sample: As used herein , the term “ sample ” refers to a to grow , expand or increase or cause to grow , expand or subset of its tissues , cells or component parts ( e.g. body increase rapidly. “ Proliferative ” means having the ability to fluids, including but not limited to peripheral blood , serum , proliferate. “ Anti -proliferative ” means having properties plasma, ascites, urine , cerebrospinal fluid ( CSF ) , sputum , counter to or inapposite to proliferative properties. saliva , bone marrow , synovial fluid , aqueous humor, amni Pharmaceutically acceptable : The phrase “ pharmaceuti- 60 otic fluid , cerumen , breast milk , broncheoalveolar lavage cally acceptable” is employed herein to refer to those fluid, semen , prostatic fluid , cowper's fluid or pre- ejacula compounds, materials, compositions , and / or dosage forms tory fluid , sweat, fecal matter, hair, tears , cyst fluid , pleural which are , within the scope of sound medical judgment, and peritoneal fluid, pericardial fluid , lymph , chyme, chyle, suitable for use in contact with the tissues of human beings bile , interstitial fluid , menses , pus , sebum , vomit , vaginal and animals without excessive toxicity, irritation , allergic 65 secretions, mucosal secretion , stool water, pancreatic juice , response , or other problem or complication , commensurate lavage fluids from sinus cavities , bronchopulmonary aspi with a reasonable benefit/ risk ratio . rates, blastocyl cavity fluid , and umbilical cord blood ). A US 10,898,574 B2 53 sample further may include a homogenate, lysate or extract prepared from a whole organism or a subset of its tissues , cells or component parts, or a fraction or portion thereof, including but not limited to , for example , plasma , serum , spinal fluid , lymph fluid , the external sections of the skin , respiratory, intestinal, and genitourinary tracts, tears, saliva , milk , blood cells , tumors, organs. A sample further refers to a medium , such as a nutrient broth or gel , which may contain cellular components, such as proteins or nucleic acid molecule . Similarity: As used herein , the term " similarity ” refers to the overall relatedness between polymeric molecules , e.g. between polynucleotide molecules ( e.g. DNA molecules and / or RNA molecules ) and / or between polypeptide molecules . Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art. Stable : As used herein “ stable ” refers to a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture , and preferably capable of formulation into an efficacious therapeutic agent. Subject: As used herein , the term “ subject ” or “ patient ” refers to any organism to which a composition in accordance with the invention may be administered , e.g. , for experimental, diagnostic, prophylactic, and / or therapeutic purposes . Typical subjects include animals (e.g. , mammals such as mice , rats, rabbits , non -human primates, and humans) and / or plants . Substantially: As used herein , the term " substantially " refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and /or proceed to completeness or achieve or avoid an absolute result . The term “ substantially ” is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical Substantially equal: As used herein as it relates to time differences between doses, the term means plus/minus 2 % . Substantially simultaneously : As used herein and as it phenomena. relates to plurality of doses , the term means within 2 seconds. Simultaneously: As used herein , “ simultaneously ” means within scientific reproducibility, at same time . Suffering from : An individual who is “ suffering from ” a or displays one or more symptoms of a disease, disorder, disease , disorder, and / or condition has been diagnosed with and / or condition . 5 10 15 20 25 30 54 ( 6 ) exposure to and /or infection with a microbe associated with development of the disease , disorder, and / or condition . In some embodiments, an individual who is susceptible to a disease , disorder, and / or condition will develop the disease , disorder, and / or condition . In some embodiments , an indi vidual who is susceptible to a disease , disorder, and /or condition will not develop the disease, disorder, and / or condition . Synthetic: The term “ synthetic ” means produced, pre pared, and / or manufactured by the hand ofman . Synthesis of polynucleotides or polypeptides or other molecules of the present invention may be chemical or enzymatic . Single unit dose : As used herein , a “ single unit dose ” is a dose of any therapeutic administered in one dose/at one time/ single route/ single point of contact, i.e. , single admin istration event. Total daily dose: As used herein , a “ total daily dose” is an amount given or prescribed in 24 hr period. It may be administered as a single unit dose . Split dose: As used herein , a “ split dose ” is the division of single unit dose or total daily dose into two or more doses. Targeted Cells : As used herein , “ targeted cells ” refers to any one or more cells of interest. The cells may be found in vitro, in vivo , in situ or in the tissue or organ of an organism . The organism may be an animal , preferably a mammal, more preferably a human and most preferably a patient. Therapeutic Agent: The term “ therapeutic agent ” refers to any agent that, when administered to a subject, has a therapeutic, diagnostic , and / or prophylactic effect and / or elicits a desired biological and / or pharmacological effect. Therapeutically effective amount: As used herein , the term “ therapeutically effective amount ” means an amount of an agent to be delivered (e.g. , nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is 35 sufficient, when administered to a subject suffering from or susceptible to a disease , disorder, and / or condition , to treat, improve symptoms of, diagnose , prevent, and / or delay the onset of the disease , disorder, and / or condition . Transcription factor: As used herein, the term “ transcrip 40 tion factor” refers to a DNA -binding protein that regulates transcription of DNA into RNA, for example , by activation or repression of transcription . Some transcription factors effect regulation of transcription alone , while others act in concert with other proteins. Some transcription factor can 45 both activate and repress transcription under certain condi tions . In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcrip tion factors may regulate transcription of a target gene alone 50 or in a complex with other molecules . Treating: As used herein , the term “ treating” refers to partially or completely alleviating, ameliorating, improving , relieving, delaying onset of, inhibiting progression of, reducing severity of, and / or reducing incidence of one or 55 more symptoms or features of a particular disease , disorder, and / or condition . For example , " treating " cancer may refer to inhibiting survival, growth , and / or spread of a tumor. Susceptible to : An individual who is " susceptible to ” a disease , disorder, and / or condition has not been diagnosed with and / or may not exhibit symptoms of the disease , disorder, and /or condition but harbors a propensity to develop a disease or its symptoms. In some embodiments, an individual who is susceptible to a disease , disorder, and / or condition ( for example, cancer) may be characterized by one Treatment may be administered to a subject who does not or more of the following: ( 1 ) a genetic mutation associated exhibit signs of a disease , disorder, and / or condition and /or with development of the disease, disorder, and / or condition ; 60 to a subject who exhibits only early signs of a disease, ( 2 ) a genetic polymorphism associated with development of disorder, and / or condition for the purpose of decreasing the the disease , disorder, and / or condition ; (3 ) increased and / or risk of developing pathology associated with the disease , decreased expression and / or activity of a protein and / or disorder, and / or condition . nucleic acid associated with the disease , disorder, and /or Unmodified : As used herein , “ unmodified ” refers to any condition ; (4 ) habits and /or lifestyles associated with devel- 65 substance, compound or molecule prior to being changed in opment of the disease , disorder, and / or condition ; ( 5 ) a any way. Unmodified may , but does not always , refer to the family history of the disease , disorder, and / or condition ; and wild type or native form of a biomolecule . Molecules may US 10,898,574 B2 55 56 undergo a series of modifications whereby each modified information available to the applicants and does not consti molecule may serve as the “ unmodified ” starting molecule tute any admission as to the correctness of the dates or for a subsequent modification. contents of these documents. All cited sources, for example, references, publications, Equivalents and Scope 5 databases, database entries, and art cited herein , are incor porated into this application by reference , even if not Those skilled in the art will recognize , or be able to expressly stated in the citation . In case of conflicting state ascertain using no more than routine experimentation, many of a cited source and the instant application, the equivalents to the specific embodiments in accordance with ments statement in the instant application shall control. the invention described herein . The scope of the present 10 invention is not intended to be limited to the above Descrip EXAMPLES tion, but rather is as set forth in the appended claims . In the claims , articles such as “ a , ” “ an ,” and “ the ” may Example 1. Modified mRNA Production mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descrip- 15 Modified mRNAs (mmRNA ) according to the invention tions that include “ or” between one or more members of a using standard laboratory methods and mate group are considered satisfied if one , more than one , or all may. beThemade open reading frame (ORF ) of the gene of interest of the group members are present in, employed in , or rials otherwise relevant to a given product or process unless may be flanked by a 5 ' untranslated region (UTR ) which may indicated to the contrary or otherwise evident from the 20 contain a strong Kozak translational initiation signal and/or context. The invention includes embodiments in which an alpha - globin 3 ' UTR which may include an oligo (dT ) exactly one member of the group is present in , employed in , sequence for templated addition of a poly - A tail . The modi or otherwise relevant to a given product or process. The fied mRNAs may be modified to reduce the cellular innate invention includes embodiments in which more than one , or immune response . The modifications to reduce the cellular all of the group members are present in , employed in , or 25 response may include pseudouridine (4 ) and 5-methyl otherwise relevant to a given product or process . cytidine ( 5meC or mC ). ( see , Kariko K et al . Immunity It is also noted that the term “ comprising " is intended to 23 : 165-75 (2005 ) , Kariko K et al . Mol Ther 16 : 1833-40 be open and permits the inclusion of additional elements or (2008 ) , Anderson B R et al . NAR ( 2010 ) ; herein incorpo steps . rated reference ). Where ranges are given, endpoints are included . Further- 30 ThebyORF may also include various upstream or down more , it is to be understood that unless otherwise indicated stream additions ( such as , but not limited to , ß - globin , tags , or otherwise evident from the context and understanding of etc. ) may be ordered from an optimization service such as , one of ordinary skill in the art, values that are expressed as but limited to , DNA2.0 ( Menlo Park , Calif.) and may ranges can assume any specific value or subrange within the contain multiple cloning sites which may have Xbal recog stated ranges in different embodiments of the invention , to 35 nition . Upon receipt of the construct, it may be reconstituted the tenth of the unit of the lower limit of the range, unless and transformed into chemically competent E. coli. the context clearly dictates otherwise . For the present invention , NEB DH5 - alpha Competent E. In addition , it is to be understood that any particular coli are used . Transformations are performed according to embodiment of the present invention that falls within the prior art may be explicitly excluded from any one or more 40 NEB instructions using 100 ng of plasmid . The protocol is : of the claims . Since such embodiments are deemed to be as 1.follows Thaw a tube ofNEB 5 - alpha Competent E. coli cells on known to one of ordinary skill in the art, they may be ice for 10 minutes. excluded even if the exclusion is not set forth explicitly herein . Any particular embodiment of the compositions of 2. Add 1-5 ul containing 1 pg - 100 ng of plasmid DNA to the invention ( e.g. , any nucleic acid or protein encoded 45 the cell mixture . Carefully flick the tube 4-5 times to mix cells and DNA . Do not vortex . thereby ; any method of production; any method of use ; etc.) can be excluded from any one or more claims, for any 3. Place the mixture on ice for 30 minutes. Do not mix . reason , whether or not related to the existence of prior art. 4. Heat shock at 42 ° C. for exactly 30 seconds . Do not As used herein and in the claims , the singular forms mix . include the plural reference and vice versa unless the context 50 5. Place on ice for 5 minutes . Do not mix . clearly indicates otherwise . Other than in the operating 6. Pipette 950 ul of room temperature SOC into the examples, or where otherwise indicated , all numbers mixture. expressing quantities of ingredients or reaction conditions 7. Place at 37 ° C. for 60 minutes. Shake vigorously ( 250 used herein should be understood as modified in all instances by the term “ about.” 55 rpm ) or rotate . 8. Warm selection plates to 37 ° C. 9. Mix the cells thoroughly by flicking the tube and inverting All patents, oligonucleotide sequences identified by gene identification numbers, and other publications identified herein are expressly incorporated by reference for the pur 10. Spread 50-100 ul of each dilution onto a selection pose of describing and disclosing , for example, the meth plate and incubate overnight at 37 ° C. Alternatively, odologies described in such publications that might be used 60 incubate at 30 ° C. for 24-36 hours or 25 ° C. for 48 in connection with the present invention . These publications hours . are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be A single colony is then used to inoculate 5 ml of LB construed as an admission that the inventors are not entitled growth media using the appropriate antibiotic and then to antedate such disclosure by virtue of prior invention or for 65 allowed to grow ( 250 RPM , 37 ° C. ) for 5 hours . This is then any other reason . All statements as to the date or represen used to inoculate a 200 ml culture medium and allowed to tation as to the contents of these documents is based on the grow overnight under the same conditions . US 10,898,574 B2 58 57 To isolate the plasmid ( up to 850 ug ), a maxi prep is Calif.) per manufacturer's instructions. Larger scale purifi performed using the Invitrogen PURELINKTM HiPure cations may need to be done with a product that has a larger Maxiprep Kit (Carlsbad, Calif. ), following the manufactur- load capacity such as Invitrogen's standard PURELINKTM er's instructions . PCR Kit ( Carlsbad , Calif .). Following the cleanup, the In order to generate cDNA for In Vitro Transcription 5 linearized vector is quantified using the NanoDrop and ( IVT ) , the plasmid (an Example of which is shown in FIG . analyzed to confirm linearization using agarose gel electro 2 ) is first linearized using a restriction enzyme such as Xbal . phoresis. A typical restriction digest with Xbal will comprise the As a non -limiting example, G -CSF may represent the following: Plasmid 1.0 ug ; 10x Buffer 1.0 ul; Xbal 1.5 ul; polypeptide of interest . Sequences used in the steps outlined dH20 up to 10 ul ; incubated at 37 ° C. for 1 hr. If performing 10 in Examples 1-5 are shown in Table 2. It should be noted that at lab scale ( < 5 ug) , the reaction is cleaned up using Invitrogen's PURELINKTM PCR Micro Kit (Carlsbad, the start codon (ATG ) has been underlined in each sequence of Table 2 . TABLE 2 G - CSF Sequences SEQ ID NO 1 Description cDNAsequence : ATGGCTGGACCTGCCACCCAGAGCCCCATGAAGCTGATGGCCCTGCAGCTGCT GCTGTGGCACAGTGCACTCTGGACAGTGCAGGAAGCCACCCCCCTGGGCCCTG CCAGCTCCCTGCCCCAGAGCTTCCTGCTCAAGTGCTTAGAGCAAGTGAGGAAG ATCCAGGGCGATGGCGCAGCGCTCCAGGAGAAGCTGGTGAGTGAGTGTGCCAC CTACAAGCTGTGCCACCCCGAGGAGCTGGTGCTGCTCGGACACTCTCTGGGCA TCCCCTGGGCTCCCCTGAGCAGCTGCCCCAGCCAGGCCCTGCAGCTGGCAGGC TGCTTGAGCCAACTCCATAGCGGCCTTTTCCTCTACCAGGGGCTCCTGCAGGCC CTGGAAGGGATCTCCCCCGAGTTGGGTCCCACCTTGGACACACTGCAGCTGGA CGTCGCCGACTTTGCCACCACCATCTGGCAGCAGATGGAAGAACTGGGAATGG CCCCTGCCCTGCAGCCCACCCAGGGTGCCATGCCGGCCTTCGCCTCTGCTTTCC AGCGCCGGGCAGGAGGGGTCCTGGTTGCCTCCCATCTGCAGAGCTTCCTGGAG GTGTCGTACCGCGTTCTACGCCACCTTGCCCAGCCCTGA 2 CDNA having T7 polymerase site and Xba restriction site : TTGGACCCTCGTACAGAAGCTAATACGACTCACTATA GGGAAATAAGAGAGAAAAGAAGAGTAAGAAGAAATATAAGAGCCACC ATGGCTGGACCTGCCACCCAGAGCCCCATGAAGCTGATGGCCCTGCAGCTGCT GCTGTGGCACAGTGCACTCTGGACAGTGCAGGAAGCCACCCCCCTGGGCCCTG CCAGCTCCCTGCCCCAGAGCTTCCTGCTCAAGTGCTTAGAGCAAGTGAGGAAG ATCCAGGGCGATGGCGCAGCGCTCCAGGAGAAGCTGGTGAGTGAGTGTGCCAC CTACAAGCTGTGCCACCCCGAGGAGCTGGTGCTGCTCGGACACTCTCTGGGCA TCCCCTGGGCTCCCCTGAGCAGCTGCCCCAGCCAGGCCCTGCAGCTGGCAGGC TGCTTGAGCCAACTCCATAGCGGCCTTTTCCTCTACCAGGGGCTCCTGCAGGCC CTGGAAGGGATCTCCCCCGAGTTGGGTCCCACCTTGGACACACTGCAGCTGGA CGTCGCCGACTTTGCCACCACCATCTGGCAGCAGATGGAAGAACTGGGAATGG CCCCTGCCCTGCAGCCCACCCAGGGTGCCATGCCGGCCTTCGCCTCTGCTTTCC AGCGCCGGGCAGGAGGGGTCCTGGTTGCCTCCCATCTGCAGAGCTTCCTGGAG GTGTCGTACCGCGTTCTACGCCACCTTGCCCAGCCCTGAAGCGCTGCCTTCTGC GGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGG TCTTTGAATAAAGCCTGAGTAGGAAGGCGGCCGCTCGAGCATGCATCTAGA 3 Optimized sequence ; containing T7 polymerase site and Xba restriction site TTGGACCCTCGTACAGAAGCTAATACGACTCACTATAGGGAAATAAGAGAGAA AAGAAGAGTAAGAAGAAATATAAGAGCCACC ATGGCCCTGCAGTTGCTGCTTTGGCACTCGGCCCTCTGGACAGTCCAAGAAGCG ACTCCTCTCGGACCTGCCTCATCGTTGCCGCAGTCATTCCTTTTGAAGTGTCTGG AGCAGGTGCGAAAGATTCAGGGCGATGGAGCCGCACTCCAAGAGAAGCTCTG CGCGACATACAAACTTTGCCATCCCGAGGAGCTCGTACTGCTCGGGCACAGCT TGGGGATTCCCTGGGCTCCTCTCTCGTCCTGTCCGTCGCAGGCTTTGCAGTTGG CAGGGTGCCTTTCCCAGCTCCACTCCGGTTTGTTCTTGTATCAGGGACTGCTGC AAGCCCTTGAGGGAATCTCGCCAGAATTGGGCCCGACGCTGGACACGTTGCAG CTCGACGTGGCGGATTTCGCAACAACCATCTGGCAGCAGATGGAGGAACTGGG GATGGCACCCGCGCTGCAGCCCACGCAGGGGGCAATGCCGGCCTTTGCGTCCG CGTTTCAGCGCAGGGCGGGTGGAGTCCTCGTAGCGAGCCACCTTCAATCATTTT TGGAAGTCTCGTACCGGGTGCTGAGACATCTTGCGCAGCCGTGAGCCTTCTGCG GGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGT CTTTGAATAAAGCCTGAGTAGGAAGGCGGCCGCTCGAGCATGCA 4 mRNA sequence ( transcribed ) CUCACUAUAGGGAAAUAAGAGAGAAAAGAAGAGUAAGAAGAAAUAUAAGAG CCACCA AUGGCCCUGCAGUUGCUGCUUUGGCACUCGGCCCUCUGGACAGUCCAAGAAG CGACUCCUCUCGGACCUGCCUCAUCGUUGCCGCAGUCAUUCCUUUUGAAGUG UCUGGAGCAGGUGCGAAAGAUUCAGGGCGAUGGAGCCGCACUCCAAGAGAA GCUCUGCGCGACAUACAAACUUUGCCAUCCCGAGGAGCUCGUACUGCUCGGG CACAGCUUGGGGAUUCCCUGGGCUCCUCUCUCGUCCUGUCCGUCGCAGGCUU UGCAGUUGGCAGGGUGCCUUUCCCAGCUCCACUCCGGUUUGUUCUUGUAUCA US 10,898,574 B2 59 60 TABLE 2 - continued G - CSF Sequences SEQ ID NO Description GGGACUGCUGCAAGCCCUUGAGGGAAUCUCGCCAGAAUUGGGCCCGACGCUG GACACGUUGCAGCUCGACGUGGCGGAUUUCGCAACAACCAUCUGGCAGCAGA UGGAGGAACUGGGGAUGGCACCCGCGCUGCAGCCCACGCAGGGGGCAAUGCC GGCCUUUGCGUCCGCGUUUCAGCGCAGGGCGGGUGGAGUCCUCGUAGCGAGC CACCUUCAAUCAUUUUUGGAAGUCUCGUACCGGGUGCUGAGACAUCUUGCG CAGCCGUGAGCCUUCUGCGGGGCUUGCCUUCUGGCCAUGCCCUUCUUCUCUC CCUUGCACCUGUACCUCUUGGUCUUUGAAUAAAGCCUGAGUAGGAAGGCGG CCGCUCGAGCAUGCAU 15 to confirm the RNA is the proper size and that no degrada Example 2 : PCR for cDNA Production PCR procedures for the preparation of cDNA are per formed using 2xKAPA HIFITM HotStart ReadyMix by Kapa Biosystems (Woburn, Mass . ) . This system includes tion of the RNA has occurred . 20 2xKAPA ReadyMix12.5 ul ; Forward Primer ( 10 uM) 0.75 ul ; Reverse Primer ( 10 uM ) 0.75 ul ; Template cDNA 100 ng; and dH2O diluted to 25.0 ul. The reaction conditions are at 95 ° C. for 5 min . and 25 cycles of 98 ° C. for 20 sec , then 58 ° 25 C. for 15 sec , then 72 ° C. for 45 sec , then 72 ° C. for 5 min . then 4 ° C. to termination . The reverse primer of the instant invention incorporates a poly - T120 for a poly - A120 in the mRNA . Other reverse primers with longer or shorter poly ( T ) tracts can be used to 30 adjust the length of the poly (A) tail in the mRNA . The reaction is cleaned up using Invitrogen’s PURE LINKTM PCR Micro Kit ( Carlsbad , Calif.) per manufactur er's instructions ( up to 5 ug ) . Larger reactions will require a cleanup using a product with a larger capacity. Following the 35 cleanup , the cDNA is quantified using the Nano Drop and analyzed by agarose gel electrophoresis to confirm the cDNA is the expected size . The cDNA is then submitted for sequencing analysis before proceeding to the in vitro tran scription reaction. Example 3. In Vitro Transcription (IVT) The in vitro transcription reaction generates mRNA con taining modified nucleotides or modified RNA . The input nucleotide triphosphate (NTP ) mix is made in -house using natural and un - natural NTPs . A typical in vitro transcription reaction includes the following: 1. Template cDNA 10x transcription buffer (400 mM Tris -HCl pH 8.0 , 190 mM MgCl2 , 50 mM DTT, 10 mm Spermidine) 2. 1.0 ug 2.0 ul 40 Example 4. Enzymatic Capping of mRNA Capping of the mRNA is performed as follows where the mixture includes : IVT RNA 60 ug - 180 ug and dH2O up to 72 ul . The mixture is incubated at 65 ° C. for 5 minutes to denature RNA, and then is transferred immediately to ice . The protocol then involves the mixing of 10x Capping Buffer ( 0.5 M Tris -HCl (pH 8.0) , 60 mM KC1 , 12.5 mM MgCl2 ) ( 10.0 ul); 20 mM GTP ( 5.0 ul ) ; 20 mM S -Adenosyl Methionine ( 2.5 ul ) ; RNase Inhibitor ( 100 U) ; 2'-O -Meth yltransferase ( 400 U) ; Vaccinia capping enzyme (Guanylyl transferase) (40 U) ; dH2O (Up to 28 ul ) ; and incubation at 37 ° C. for 30 minutes for 60 ug RNA or up to 2 hours for 180 ug of RNA . The mRNA is then purified using Ambion’s MEGA CLEARTM Kit ( Austin, Tex .) following the manufacturer's instructions. Following the cleanup, the RNA is quantified using the NANODROPTM ( ThermoFisher, Waltham , Mass . ) and analyzed by agarose gel electrophoresis to confirm the RNA is the proper size and that no degradation of the RNA has occurred . The RNA product may also be sequenced by running a reverse - transcription -PCR to generate the cDNA for sequencing 45 Example 5. PolyA Tailing Reaction Without a poly - T in the cDNA, a poly -A tailing reaction 50 must be performed before cleaning the final product. This is done by mixing Capped IVTRNA ( 100 ul ) ; RNase Inhibitor (20 U) ; 10x Tailing Buffer (0.5 M Tris - HCl ( pH 8.0 ) , 2.5 M NaCl , 100 mM MgCl2 ) ( 12.0 ul ) ; 20 mM ATP ( 6.0 ul ) ; Poly -A Polymerase ( 20 U) ; dH20 up to 123.5 ul and RNase Inhibitor 20 U 55 incubation at 37 ° C. for 30 min . If the poly -A tail is already 3000 U 5. T7 RNA polymerase in the transcript , then the tailing reaction may be skipped and 6 . dH20 Up to 20.0 ul . and proceed directly to cleanup with Ambion's MEGA 7 . Incubation at 37 ° C. for 3 hr- 5 hrs . CLEARTM kit ( Austin , Tex .) ( up to 500 ug ) . Poly - A Poly merase The crude IVT mix may be stored at 4 ° C. overnight for 60 yeast . is preferably a recombinant enzyme expressed in cleanup the next day. 1 U of RNase - free DNase is then used to digest the original template. After 15 minutes of incubaFor studies performed and described herein , the poly -A tion at 37 ° C. , the mRNA is purified using Ambion's tail is encoded in the IVT template to comprise 160 nucleo MEGACLEARTM Kit ( Austin , Tex .) following the manu- tides in length . However, it should be understood that the 3. 4. Custom NTPs (25 mM each ) 7.2 ul facturer's instructions. This kit can purify up to 500 ug of 65 processivity or integrity of the Poly-A tailing reaction may RNA . Following the cleanup , the RNA is quantified using the NanoDrop and analyzed by agarose gel electrophoresis not always result in exactly 160 nucleotides. Hence Poly -A tails of approximately 160 nucleotides, e.g , about 150-165 , US 10,898,574 B2 61 62 155 , 156 , 157 , 158 , 159 , 160 , 161 , 162 , 163 , 164 or 165 are within the scope of the invention . Example 6. Formulation of Modified mRNA Using Lipidoids D. Capping Reaction Efficiency Synthetic mRNAs encoding human G - CSF ( cDNA shown 5 in SEQ ID NO : 1 ) containing the ARCA cap analog or the Capl structure can be analyzed for capping reaction effi ciency by LC - MS after capped mRNA nuclease treatment. Nuclease treatment of capped mRNAs would yield a mix ture of free nucleotides and the capped 5 ' - 5 - triphosphate cap structure detectable by LC - MS . The amount of capped product on the LC - MS spectra can be expressed as a percent of total mRNA from the reaction and would correspond to capping reaction efficiency. The cap structure with a higher capping reaction efficiency would have a higher amount of 5 ' - capping of modified RNA may be completed concomitantly during the in vitro -transcription reaction using the following chemical RNA cap analogs to generate the 5 - guanosine cap structure according to manufacturer pro- 10 tocols : 3 ' - O - Me - m7G ( 5 ' ) ppp ( 5 ' ) G [ the ARCA cap ]; G ( 5 ' ) ppp ( 5 ' ) A ; G ( 5'ppp ( 5 ' ) G ; m7G ( 5 ') ppp ( 5 ') A ; m76 ( 5 ')ppp ( 5 ' ) G (New England BioLabs , Ipswich , Mass . ) . 5 ' - capping of modified RNA may be completed post -transcriptionally capped product by LC -MS . using a Vaccinia Virus Capping Enzyme to generate the 15 Example 8. Formulation of Modified mRNA Using “ Cap 0 ” structure: m7G ( 5 ' ) ppp ( 5 ' ) G (New England Bio Lipidoids Labs , Ipswich , Mass . ) . Cap 1 structure may be generated using both Vaccinia Virus Capping Enzyme and a 2-0 Modified mRNAs (mmRNA ) are formulated for in vitro methyl -transferase to generate: m7G (54)ppp (5 ')G -2-0 methyl. Cap 2 structure may be generated from the Cap 1 20 experiments by mixing the mmRNA with the lipidoid at a set structure followed by the 2 - O -methylation of the 5'-ante ratio prior to addition to cells . In vivo formulation may penultimate nucleotide using a 2-0 methyl - transferase . Cap require the addition of extra ingredients to facilitate circu 3 structure may be generated from the Cap 2 structure lation throughout the body. To test the ability of these followed by the 2 - O -methylation of the 5'-preantepenulti- lipidoids to form particles suitable for in vivo work , a mate nucleotide using a 2 ' -0 methyl -transferase. Enzymes 25 standard formulation process used for siRNA - lipidoid for mulations was used as a starting point. Initial mmRNA are preferably derived from a recombinant source . When transfected into mammalian cells , the modified lipidoid formulations may consist of particles composed of mRNAs have a stability of between 12-18 hours or more 42 % lipidoid, 48 % cholesterol and 10 % PEG , with further than 18 hours , e.g. , 24 , 36 , 48 , 60 , 72 or greater than 72 optimization of ratios possible . After formation of the par hours. 30 ticle , mmRNA is added and allowed to integrate with the complex. The encapsulation efficiency is determined using a Example 7. Capping standard dye exclusion assays . Materials and Methods for Examples 9-13 A. Lipid Synthesis A. Protein Expression Assay Synthetic mRNAs encoding human G - CSF ( cDNA shown 35 Six lipids , DLin - DMA , DLin- K - DMA , DLin -KC2 -DMA , in SEQ ID NO : 1 ) containing the ARCA (3 ' O - Me -m7G ( 5 ' ) 98N12-5 , C12-200 and DLin -MC3 -DMA, were synthesized ppp ( 5 ' ) G ) cap analog or the Capl structure can be trans- by methods outlined in the art in order to be formulated with fected into human primary keratinocytes at equal concen modified RNA . DLin - DMA and precursors were synthe trations. 6 , 12 , 24 and 36 hours post - transfection the amount sized as described in Heyes et. al , J. Control Release , 2005 , of G - CSF secreted into the culture medium can be assayed 40 107 , 276-287 . DLin - K - DMA and DLin - KC2 - DMA and pre by ELISA . Synthetic mRNAs that secrete higher levels of cursors were synthesized as described in Semple et . al , G - CSF into the medium would correspond to a synthetic Nature Biotechnology, 2010 , 28 , 172-176 . 98N12-5 and mRNA with a higher translationally -competent Cap struc- precursor were synthesized as described in Akinc et . al , ture . Nature Biotechnology, 2008 , 26 , 561-569 . B. Purity Analysis Synthesis 45 C12-200 and precursors were synthesized according to mRNAs encoding human G - CSF ( cDNA shown in SEQ ID NO : 1 ) containing the ARCA cap analog or the Cap1 structure crude synthesis products can be compared for purity using denaturing Agarose -Urea gel electrophoresis or the method outlined in Love et . al , PNAS , 2010 , 107 , 1864-1869 . 2 -epoxydodecane ( 5.10 g , 27.7 mmol, 8.2 eq) was added to a vial containing Amine 200 (0.723 g , 3.36 mmol, 1 eq) and a stirring bar. The vial was sealed and HPLC analysis. Synthetic mRNAs with a single, consoli- 50 warmed to 80 ° C. The reaction was stirred for 4 days at 80 ° dated band by electrophoresis correspond to the higher C. Then the mixture was purified by silica gel chromatog purity product compared to a synthetic mRNA with multiple raphy using a gradient from pure dichloromethane ( DCM ) to bands or streaking bands . Synthetic mRNAs with a single DCM : MeOH 98 : 2 . The target compound was further puri HPLC peak would also correspond to a higher purity prod- fied by RP - HPLC to afford the desired compound. uct . The capping reaction with a higher efficiency would 55 DLin -MC3 -DMA and precursors were synthesized provide a more pure mRNA population . according to procedures described in WO 2010054401 C. Cytokine Analysis herein incorporated by reference in its entirety . A mixture of Synthetic mRNAs encoding human G - CSF (cDNA shown dilinoleyl methanol ( 1.5 g , 2.8 mmol , 1 eq) , N , N -dimethyl in SEQ ID NO : 1 ) containing the ARCA cap analog or the aminobutyric acid ( 1.5 g , 2.8 mmol, 1 eq ), DIPEA (0.73 mL , Capl structure can be transfected into human primary kera- 60 4.2 mmol, 1.5 eq ) and TBTU ( 1.35 g , 4.2 mmol, 1.5 eq) in tinocytes at multiple concentrations. 6 , 12 , 24 and 36 hours 10 mL of DMF was stirred for 10 h at room temperature. post - transfection the amount of pro -inflammatory cytokines Then the reaction mixture was diluted in ether and washed such as TNF - alpha and IFN -beta secreted into the culture with water . The organic layer was dried over anhydrous medium can be assayed by ELISA . Synthetic mRNAs that sodium sulfate, filtrated and concentrated under reduced secrete higher levels of pro - inflammatory cytokines into the 65 pressure. The crude product was purified by silica gel medium would correspond to a synthetic mRNA containing chromatography using a gradient DCM to DCM : MeOH 98 : 2 . Subsequently the target compound was subjected to an an immune -activating cap structure . US 10,898,574 B2 64 63 additional RP - HPLC purification which was done using a YMC - Pack C4 column to afford the target compound. B. Formulation of Modified RNA Nanoparticles Solutions of synthesized lipid, 1,2- distearoyl-3 -phosphatidylcholine (DSPC) ( Avanti Polar Lipids , Alabaster, Ala . ) , 5 cholesterol ( Sigma - Aldrich, Taufkirchen , Germany ), and a-[ 3'- (1,2 -dimyristoyl- 3 -propanoxy)-carboxamide-propyl]0 -methoxy -polyoxyethylene ( PEG - C - DOMG) (NOF, Bouwelven , Belgium) were prepared at concentrations of 50 mM in ethanol and stored at -20 ° C. The lipids were 10 combined to yield molar ratio of 50 : 10 : 38.5 : 1.5 ( Lipid : DSPC : Cholesterol: PEG - c - DOMG) and diluted with ethanol to a final lipid concentration of 25 mM . Solutions of modified mRNA at a concentration of 1-2 mg /mL in water were diluted in 50 mM sodium citrate buffer at a pH of 3 to 15 form a stock modified mRNA solution . Formulations of the lipid and modified mRNA were prepared by combining the synthesized lipid solution with the modified mRNA solution at total lipid to modified mRNA weight ratio of 10 : 1 , 15 : 1 , 20 : 1 and 30 : 1 . The lipid ethanolic solution was rapidly 20 injected into aqueous modified mRNA solution to afford a suspension containing 33 % ethanol. The solutions were injected either manually (MI ) or by the aid of a syringe pump ( SP) (Harvard Pump 33 Dual Syringe Pump Harvard Apparatus Holliston , Mass . ) . 25 To remove the ethanol and to achieve the buffer exchange, the formulations were dialyzed twice against phosphate buffered saline (PBS ) , pH 7.4 at volumes 200 -times of the primary product using a Slide - A - Lyzer cassettes ( Thermo Fisher Scientific Inc. Rockford , Ill . ) with a molecular weight 30 cutoff ( MWCO ) of 10 kD . The first dialysis was carried at room temperature for 3 hours and then the formulations were dialyzed overnight at 4 ° C. The resulting nanoparticle suspension was filtered through 0.2 um sterile filter ( Sarstedt, Niimbrecht, Germany) into glass vials and sealed 35 with a crimp closure . C. Characterization of Formulations A Zetasizer Nano ZS (Malvern Instruments Ltd, Malvern , Worcestershire , UK) was used to determine the particle size , the polydispersity index (PDI ) and the zeta potential of the 40 modified mRNA nanoparticles in 1xPBS in determining particle size and 15 mM PBS in determining zeta potential. Ultraviolet - visible spectroscopy was used to determine the concentration of modified mRNA nanoparticle formulation. 100 uL of the diluted formulation in 1xPBS was added 45 to 900 uL of a 4 : 1 ( v / v ) mixture of methanol and chloroform . After mixing , the absorbance spectrum of the solution was recorded between 230 nm and 330 nm on a DU 800 spectrophotometer ( Beckman Coulter, Beckman Coulter, nanoparticle formulation was calculated based on the extinc Inc., Brea, Calif .). The modified RNA concentration in the 50 tion coefficient of the modified RNA used in the formulation and on the difference between the absorbance at a wave- length of 260 nm and the baseline value at a wavelength of 330 nm . QUANT-ITTM RIBOGREEN® RNA assay ( Invitrogen Corporation Carlsbad, Calif .) was used to evaluate the encapsulation of modified RNA by the nanoparticle. The samples were diluted to a concentration of approximately 5 ug /mL in TE buffer ( 10 mM Tris - HCl, 1 mM EDTA , pH 7.5 ) . 50 uL of the diluted samples were transferred to a polystyrene 96 well plate , then either 50 uL of TE buffer or 50 uL of a 2 % Triton X - 100 solution was added . The plate was incubated at a temperature of 37 ° C. for 15 minutes. The RIBOGREEN® reagent was diluted 1 : 100 in TE buffer, 100 uL of this solution was added to each well . The fluorescence intensity was measured using a fluorescence plate reader ( Wallac Victor 1420 Multilablel Counter; Perkin Elmer, Waltham , Mass . ) at an excitation wavelength of ~ 480 nm and an emission wavelength of ~ 520 nm . The fluorescence values of the reagent blank were subtracted from that of each of the samples and the percentage of free modified RNA was determined by dividing the fluorescence intensity of the intact sample (without addition of Triton X - 100 ) by the fluorescence value of the disrupted sample ( caused by the addition of Triton X - 100 ). D. In Vitro Incubation Human embryonic kidney epithelial (HEK293 ) and hepa tocellular carcinoma epithelial (HepG2 ) cells (LGC stan dards GmbH , Wesel, Germany ) were seeded on 96 - well plates (Greiner Bio - one GmbH , Frickenhausen , Germany ) and plates for HEK293 cells were precoated with collagen typel. HEK293 were seeded at a density of 30,000 and HepG2 were seeded at a density of 35,000 cells per well in 100 ul cell culture medium . For HEK293 the cell culture medium was DMEM , 10 % FCS , adding 2 mM L - Glutamine , 1 mM Sodiumpyruvate and 1x non - essential amino acids ( Biochrom AG , Berlin , Germany) and 1.2 mg/ml Sodium bicarbonate ( Sigma - Aldrich, Munich, Germany ) and for HepG2 the culture medium was MEM (Gibco Life Tech nologies , Darmstadt, Germany ), 10% FCS adding 2 mM L -Glutamine, 1 mM Sodiumpyruvate and 1x non -essential amino acids (Biochrom AG , Berlin , Germany. Formulations containing mCherry mRNA (mRNA sequence shown in SEQ ID NO : 5 ; poly -A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl) were added in qua druplicates directly after seeding the cells and incubated . The mCherry cDNA with the T7 promoter, 5'untranslated region (UTR ) and 3 ' UTR used in in vitro transcription (IVT ) is given in SEQ ID NO : 6 . Cells were harvested by transferring the culture media supernatants to a 96 -well Pro -Bind U - bottom plate (Beckton Dickinson GmbH , Heidelberg , Germany ). Cells were trypsinized with 1/2 volume Trypsin /EDTA ( Biochrom AG , Berlin, Germany), pooled with respective supernatants and fixed by adding one volume PBS /2 % FCS (both Biochrom AG, Berlin, Germany) /0.5% formaldehyde (Merck, Darm stadt, Germany ). Samples then were submitted to a flow cytometer measurement with a 532 nm excitation laser and the 610/20 filter for PE - Texas Red in a LSRII cytometer (Beckton Dickinson GmbH , Heidelberg , Germany ). The mean fluorescence intensity (MFI ) of all events and the standard deviation of four independent wells are presented in for samples analyzed . Example 9. Purification on Nanoparticle Formulations Nanoparticle formulations of DLin -KC2 -DMA and 98N12-5 in HEK293 and HepG2 were tested to determine if the mean fluorescent intensity (MFI ) was dependent on the 55 lipid to modified RNA ratio and / or purification. Three for mulations of DLin -KC2 - DMA and two formulations of 98N12-5 were produced using a syringe pump to the speci fications described in Table 3. Purified samples were purified by SEPHADEXTM G - 25 DNA grade (GE Healthcare , Swe 60 den ). Each formulation before and after purification (aP ) were tested at concentration of 250 ng modified RNA per well in a 24 well plate . The percentage of cells that are positive for the marker for FL4 channel (% FL4 -positive ) when analyzed by the flow cytometer for each formulation 65 and the background sample are shown in FIGS . 3A and 3B , and the MFI of the marker for the FL4 channel for each formulation and the background sample are shown in FIGS . US 10,898,574 B2 65 66 4A and 4B . The formulations which had been purified had a purification. TABLE 4 slightly higher MFI than those formulations tested before Formulations Formulation # 5 TABLE 3 Formulations Formulation # Lipid 10 NPA -001-1 aP DLin -KC2 - DMA 10 NPA -002-1 DLin -KC2 -DMA 15 NPA -002-1 aP DLin -KC2- DMA 15 125 nm NPA -003-1 DLin - KC2 - DMA 20 PDI : 0.12 114 nm PDI : 0.08 NPA -003-1 aP DLin -KC2 -DMA 20 155 nm NPA - 005 DLin -KC2 98N12-5 Lipid / RNA 20 15 114 nm PDI : 0.08 106 nm PDI : 0.12 DMA Lipid / RNA wt/ wt Mean size (nm) DLin -KC2 - DMA NPA -001-1 NPA - 003 Lipid wt/wt Mean size 10 PDI : 0.08 141 nm PDI : 0.14 140 nm PDI : 0.11 TABLE 5 15 Concentration and MFI 104 nm MFI mCherry PDI : 0.06 NPA - 005-1 98N12-5 15 127 nm PDI : 0.12 NPA - 005-1 aP 98N12-5 15 98N12 20 NPA -006-1 aP 98N12 20 NPA - 006-1 Formulation 20 134 nm PDI : 0.17 126 nm PDI : 0.08 118 nm NPA - 003 NPA - 005 25 ngwell 11963.25 0.25 ng /well 0.025 ng /well 1349.75 459.50 310.75 12256.75 2572.75 534.75 471.75 0.0025 ng/well 25 PDI : 0.13 Example 11. Manual Injection and Syringe Pump Formulations Example 10. Concentration Response Curve Nanoparticle formulations of 98N12-5 (NPA -005 ) and trations to determine the MFI of FL4 or mCherry (mRNA sequence shown in SEQ ID NO : 5 ; poly -A tail of approxi 30 DLin - KC2 - DMA (NPA - 003 ) were tested at varying concen Two formulations of DLin -KC2 -DMA and 98N12-5 were prepared by manual injection ( MI ) and syringe pump injec tion ( SP ) and analyzed along with a background sample to compare the MFI of mCherry (mRNA shown in SEQ ID NO : 5 ; poly -A tail of approximately 160 nucleotides not 35 mately 160 nucleotides not shown in sequence; 5 ' cap , Capl) over a range of doses. The formulations tested are outlined in Table 4. To determine the optimal concentration of nanoparticle formulations of 98N12-5 , varying concentra 40 tions of formulated modified RNA ( 100 ng , 10 ng , 1.0 ng , 0.1 ng and 0.01 ng per well ) were tested in a 24 -well plate of HEK293 , and the results of the FL4 MFI of each dose are shown in FIG . 5A . Likewise, to determine the optimal concentration of nanoparticle formulations of DLin -KC2 45 DMA, varying concentrations of formulated modified RNA (250 ng 100 ng, 10 ng , 1.0 ng , 0.1 ng and 0.01 ng per well) were tested in a 24 -well plate of HEK293 , and the results of the FL4 MFI of each dose are shown in FIG . 5B . Nanopar shown in sequence ; 5 ' cap , Capl) of the different formula tions . Table 5 shows that the syringe pump formulations had a higher MFI as compared to the manual injection formu lations of the same lipid and lipid / RNA ratio . TABLE 5 Formulations and MFI Lipid / Formulation # Lipid wt/wt (nm ) Untreated Control NPA -002 N/A N /A DLin -KC2 15 DLin -KC2 varying concentrations of formulated modified RNA (250 NPA - 003 DMA DLin -KC2 DMA 20 and the results of the FL4 MFI of each dose are shown in NPA - 003-2 DLin -KC2 20 FIG . 5C . A dose of 1 ng/well for 98N12-5 and a dose of 10 ng /well for DLin - K2 - DMA were found to resemble the FL4 55 NPA - 005 DMA 98N12-5 NPA - 005-2 98N12-5 15 To determine how close the concentrations resembled the NPA - 006 98N12-5 20 ng, 100 ng and 30 ng per well ) in a 24 well plate of HEK293 , MFI of the background. background, we utilized a flow cytometer with optimized filter sets for detection of mCherry expression, and were able 60 to obtain results with increased sensitivity relative to back ground levels . Doses of 25 ng/well, 0.25 ng /well, 0.025 ng /well and 0.0025 ng/well were analyzed for 98N12-5 (NPA -005 ) and DLin -K2 - DMA ( NPA - 003) to determine the MFI of mCherry. As shown in Table 5 , the concentration of 65 0.025 ng/well and lesser concentrations are similar to the background MFI level of mCherry which is about 386.125 . for mulation MFI 674.67 N/A N /A 140 nm MI 10318.25 SP 37054.75 MI 22037.5 SP 37868.75 MI 11504.75 SP 9343.75 MI 11182.25 SP 5167 PDI : 0.11 DMA NPA - 002-2 ticle formulations of DLin -KC2 -DMA were also tested at 50 Method of RNA Mean size 15 105 nm PDI : 0.04 114 nm PDI : 0.08 95 nm PDI : 0.02 15 127 nm PDI : 0.12 106 nm PDI : 0.07 126 nm PDI : 0.08 NPA -006-2 98N12-5 20 93 nm PDI : 0.08 Example 12. mCherry Fluorescence of Formulations Formulations of DLin - DMA , DLin - K - DMA , DLin -KC2 DMA , 98N12-5 , C12-200 and DLin -MC3 -DMA were incu US 10,898,574 B2 67 68 bated at a concentration of 60 ng /well or 62.5 ng /well in a administered the formulation . Serum from the mice admin plate of HEK293 and 62.5 ng /well in a plate of HepG2 cells for 24 hours to determine the MFI of mCherry (mRNA istered formulations containing human G - CSF modified mRNA are measured by specific G - CSF ELISA and serum shown in SEQ ID NO : 5 ; poly -A tail of approximately 160 from mice administered human Factor IX modified RNA is nucleotides not shown in sequence ; 5 ' cap , Capl ) for each 5 analyzed by specific Factor IX ELISA or chromogenic assay. The liver and spleen from the mice administered with formulation . The formulations tested are outlined in Table 6 mCherry modified mRNA are analyzed by immunohisto chemistry ( IHC ) or fluorescence - activated cell sorting ( FACS ). As a control, a group of mice are not injected with below. As shown in FIG . 6A for the 60 ng /well and FIGS . 6B , 6C , 6D , and 6E for the 62.5 ng /well, the formulation of NPA -003 and NPA -018 have the highest mCherry MFI and 10 any formulation and their serum and tissue are collected the formulations of NPA -008, NPA -010 and NPA -013 are most the similar to the background sample mCherry MFI analyzed by ELISA , FACS and / or IHC . value . Example 14. In Vitro and In Vivo Expression TABLE 6 Formulation # Lipid DLin - KC2 - DMA NPA -001 Formulations Lipid / RNA wt/wt Mean size (nm) 10 155 nm PDI : 0.08 NPA - 002 DLin -KC2 - DMA 15 NPA - 002-2 DLin -KC2 - DMA 15 105 nm PDI : 0.04 114 nm PDI : 0.08 NPA - 003 DLin -KC2 -DMA 20 NPA -003-2 DLin -KC2 - DMA 20 140 nm PDI : 0.11 95 nm PDI : 0.02 NPA - 005 98N12-5 15 127 nm PDI : 0.12 NPA - 006 98N12-5 A. A. In Vitro Expression in Human Cells Using Lipidoid The ratio of mmRNA to lipidoid used to test for in vitro transfection is tested empirically at different lipidoid : mmRNA ratios. Previous work using siRNA and lipidoids 20 have utilized 2.5 : 1 , 5 : 1 , 10 : 1 , and 15 : 1 lipidoid : siRNA wt:wt ratios. Given the longer length of mmRNA relative to siRNA , a lower wt : wt ratio of lipidoid to mmRNA may be effective. In addition, for comparison mmRNA were also formulated using RNAIMAXTM ( Invitrogen, Carlsbad , Calif.) or TRANSIT -mRNA (Mirus Bio , Madison , Wis .) 25 cationic lipid delivery vehicles . The ability of lipidoid formulated Luciferase (IVT cDNA sequence as shown in 15 Formulations 20 126 nm PDI : 0.08 NPA -007 DLin - DMA 15 NPA - 008 DLin - K - DMA 15 NPA -009 C12-200 15 NPA -010 DLin - MC3 - DMA 15 NPA -012 DLin - DMA 20 NPA -013 DLin- K- DMA 20 104 nm PDI : 0.03 101 nm PDI : 0.06 148 nm PDI : 0.09 121 nm PDI : 0.08 NPA -014 C12-200 20 NPA -015 DLin- MC3 - DMA 20 138 nm PDI : 0.15 126 nm PDI : 0.09 86 nm PDI : 0.08 109 nm PDI : 0.07 SEQ ID NO : 10) , green fluorescent protein (GFP ) ( IVT cDNA sequence as shown in SEQ ID NO : 11 ) , G- CSF (mRNA sequence shown in SEQ ID NO : 4 ; poly -A tail of 30 approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl ) , and EPO mmRNA (mRNA sequence shown in SEQ ID NO : 7 ; poly -A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl ) to express the desired protein product can be confirmed by luminescence for expression, 35 luciferase expression, flow cytometry for GFP and by ELISA for G - CSF and Erythropoietin ( EPO ) secre tion . B. In Vivo Expression Following Intravenous Injection Systemic intravenous administration of the formulations are created using various different lipidoids including, but 40 not limited to , 98N12-5 , C12-200 , and MD1 . Lipidoid formulations containing mmRNA are injected 45 Example 13. In Vivo Formulation Studies intravenously into animals . The expression of the modified mRNA (mmRNA ) -encoded proteins are assessed in blood and / or other organs samples such as , but not limited to , the liver and spleen collected from the animal . Conducting single dose intravenous studies will also allow an assess ment of the magnitude, dose responsiveness, and longevity of expression of the desired product. In one embodiment, lipidoid based formulations of Mice ( n = 5 ) are administered intravenously a single dose of a formulation containing a modified mRNA and a lipid . 98N12-5 , C12-200 , MD1 and other lipidoids, are used to The modified mRNA administered to the mice is selected 50 deliver luciferase, green fluorescent protein (GFP ), mCherry from G - CSF (mRNA shown in SEQ ID NO : 4 ; poly - A tail fluorescent protein , secreted alkaline phosphatase ( SAP) , of approximately 160 nucleotides not shown in sequence ; 5 ' human G - CSF, human Factor IX , or human Erythropoietin cap , Capl ) , erythropoietin (EPO ) (mRNA shown in SEQ ID (EPO ) mmRNA into the animal. After formulating mmRNA NO : 7 ; poly -A tail of approximately 160 nucleotides not with a lipid , as described previously, animals are divided shown in sequence ; 5 ' cap , Capl ) , Factor IX (mRNA shown 55 into groups to receive either a saline formulation, or a in SEQ ID NO : 8 ; poly - A tail of approximately 160 nucleo- lipidoid - formulation which contains one of a different selected from luciferase, GFP , mCherry, SAP , tides not shown in sequence ; 5 ' cap , Capl ) or mCherry mmRNA G - CSF, human Factor IX , and human EPO . Prior to (mRNA sequence shown in SEQ ID NO : 5 ; poly - A tail of human injection into the animal , mmRNA -containing lipidoid for approximately 160 nucleotides not shown in sequence ; 5 ' mulations are diluted in PBS . Animals are then administered cap , Capl ) . The erythropoietin cDNA with the T7 promoter, 60 a single dose of formulated mmRNA ranging from a dose of 5'untranslated region (UTR ) and 3 ' UTR used in in vitro 10 mg/kg to doses as low as 1 ng/kg, with a preferred range transcription (IVT) is given in SEQ ID NO : 9 . to be 10 mg / kg to 100 ng /kg, where the dose of mmRNA Each formulation also contains a lipid which is selected depends on the animal body weight such as a 20 gram mouse from one of DLin - DMA , DLin - K - DMA, DLin -KC2 -DMA , receiving a maximum formulation of 0.2 ml ( dosing is based 98N12-5, C12-200 or DLin-MC3- DMA. The mice are 65 no mmRNA per kg body weight).After the administration of injected with 100 ug , 10 ug or 1 ug of the formulated the mmRNA - lipidoid formulation, serum , tissues , and / or modified mRNA and are sacrificed 8 hours after they are tissue lysates are obtained and the level of the mmRNA US 10,898,574 B2 69 70 encoded product is determined at a single and /or a range of mCherry fluorescent protein, secreted alkaline phosphatase time intervals . The ability of lipidoid - formulated Luciferase , ( SAP ) , human G - CSF, human factor IX , or human Erythro GFP, mCherry, SAP, G - CSF, Factor IX , and EPO mmRNA poietin ( EPO ) mmRNA to express the desired protein prod to express the desired protein product is confirmed by uct is confirmed by luminescence for luciferase expression, luminescence for the expression of Luciferase, flow cytom- 5 flow cytometry for GFP and mCherry expression , by enzy etry for the expression of GFP and mCherry expression , by matic activity for SAP, and by ELISA for G - CSF, Factor IX enzymatic activity for SAP, or by ELISA for the section of and Erythropoietin ( EPO ) secretion . G - CSF, Factor IX and / or EPO . Additional studies for a multi-dose regimen are also Further studies for a multi -dose regimen are also per formed to determine the maximal expression of mmRNA, to performed to determine the maximal expression using evaluate the saturability of the mmRNA - driven expression 10 mmRNA, to evaluate the saturability of the mmRNA - driven (by giving a control and active mmRNA formulation in expression (achieved by giving a control and active parallel or in sequence ), and to determine the feasibility of mmRNA formulation in parallel or in sequence ), and to repeat drug administration (by giving mmRNA in doses determine the feasibility of repeat drug administration (by separated by weeks ormonths and then determining whether giving mmRNA in doses separated by weeks or months and expression level is affected by factors such as immunoge- 15 then determining whether expression level is affected by nicity) . An assessment of the physiological function of factors such as immunogenicity ) . Studies utilizing multiple proteins such as G -CSF and EPO are also determined subcutaneous or intramuscular injection sites at one time through analyzing samples from the animal tested and point , are also utilized to further increase mmRNA drug detecting increases in granulocyte and red blood cell counts, exposure and improve protein production. An assessment of respectively. Activity of an expressed protein product such 20 the physiological function of proteins , such as GFP, as Factor IX , in animals can also be assessed through mCherry , SAP, human G - CSF, human factor IX , and human analysis of Factor IX enzymatic activity ( such as an acti EPO , are determined through analyzing samples from the vated partial thromboplastin time assay ) and effect of clot tested animals andcounts detecting a change in granulocyte and /or ting times . red blood cell . Activity of an expressed protein C. In Vitro Expression Following Intramuscular and / or 25 product such as Factor IX , in animals can also be assessed Subcutaneous Injection through analysis of Factor IX enzymatic activity ( such as an The use of lipidoid formulations to deliver oligonucle activated partial thromboplastin time assay) and effect of otides, including mRNA , via an intramuscular route or a clotting times . subcutaneous route of injection needs to be evaluated as it has not been previously reported. Intramuscular and / or sub Example 15. Split Dose Studies cutaneous injection of mmRNA are evaluated to determine 30 if mmRNA -containing lipidoid formulations are capable to produce both localized and systemic expression of a desired Studies utilizing multiple subcutaneous or intramuscular portions. injection at one time point were designed and per Lipidoid formulations of 98N12-5 , C12-200 , and MD1 formed to sites investigate ways to increase mmRNA drug expo containing mmRNA selected from luciferase , green fluores cent protein (GFP) , mCherry fluorescent protein, secreted 35 sure andtheimprove protein production to detec of expressed protein product., In an addition assessment of the alkaline phosphatase (SAP ), human G - CSF, human factor tion physiological function of proteins was also determined IX , or human Erythropoietin ( EPO ) mmRNA are injected through analyzing samples from the animal tested . intramuscularly and / or subcutaneously into animals. The Surprisingly , it has been determined that split dosing of expression of mmRNA - encoded proteins are assessed both within the muscle or subcutaneous tissue and systemically in 40 mmRNA produces greater protein production and pheno blood and other organs such as the liver and spleen . Single multi typic -responses than those produced by single unit dosing or dosing schemes . dose studies allow an assessment of the magnitude, dose responsiveness, and longevity of expression of the desired The design of a single unit dose , multi -dose and split dose product. experiment involved using human erythropoietin (EPO ) Animals are divided into groups to receive either a saline 45 mmRNA (mRNA sequence shown in SEQ ID NO : 7 ; poly -A formulation or a formulation containing modified mRNA . tail of approximately 160 nucleotides not shown in Prior to injection mmRNA -containing lipidoid formulations sequence; 5 ' cap , Capl ) administered in buffer alone . The are diluted in PBS . Animals are administered a single intramuscular dose of formulated mmRNA ranging from 50 mg/kg to doses as low as 1 ng /kg with a preferred range to dosing vehicle (F. buffer) consisted of 150 mM NaCl , 2 mM CaCl2, 2 mM Nat -phosphate ( 1.4 mM monobasic sodium phosphate; 0.6 mM dibasic sodium phosphate ), and 0.5 mm be 10 mg/kg to 100 ng /kg. A maximum dose for intramus- 50 EDTA, pH 6.5. The pH was adjusted using sodium hydrox cular administration , for a mouse , is roughly 1 mg mmRNA ide and the final solution was filter sterilized . The mmRNA or as low as 0.02 ng mmRNA for an intramuscular injection was modified with 5meC at each cytosine and pseudouridine into the hind limb of the mouse . For subcutaneous admin replacement at each uridine site . istration , the animals are administered a single subcutaneous Animals ( n = 5 ) were injected IM ( intramuscular ) for the dose fromrange 400 mg kg to80 55 single unitdose of 100 ug. For multi-dosing, two schedules doses ofasformulated low as 1 ngmmRNA /kg withranging a preferred to /be were used doses of 100 ug and 6 doses of 100 ug . For the mg/kg to 100 ng/kg. A maximum dose for subcutaneous split dosing, 3 scheme schedules were used , 3 doses at administration, for a mouse , is roughly 8 mg mmRNA or as 33.3 ug and 6 doses, two of 16.5 ug mmRNA . Control dosing low as 0.02 ng mmRNA . involved use of buffer only at 6 doses. Control mmRNA For a 20 gram mouse the volume of a single intramuscular the use of luciferase mmRNA ( IVT cDNA injection is maximally 0.025 ml and a single subcutaneous 60 involved sequence shown in SEQ ID NO : 10) dosed 6 times at 100 ug . injection is maximally 0.2 ml . The optimal dose of mmRNA Blood administered is calculated from the body weight of the tion . and muscle tissue were evaluated 13 hrs post injec animal. At various points in time points following the Human EPO protein was measured in mouse serum 13 h administration of the mmRNA - lipidoid, serum , tissues , and tissue lysates is obtained and the level of the mmRNA- 65 post I.M. single , multi- or split dosing of the EPO mmRNA encoded product is determined. The ability of lipidoid- in buffer. Seven groups of mice ( n = 5 mice per group ) were formulated luciferase, green fluorescent protein ( GFP ) , treated and evaluated . The results are shown in Table 7 . US 10,898,574 B2 72 71 TABLE 7 Split dose study Group Treatment 1 Human EPO mmRNA 2 3 4 5 6 7 Human EPO mmRNA Human EPO mmRNA Human EPO mmRNA Human EPO mmRNA Luciferase mmRNA Buffer Alone Avg. Polypeptide pmol/mL per unit (pmol /ug ) Dose of mmRNA Total Dose human EPO 1 x 100 ug 3 x 100 ug 6 x 100 ug 3 x 33.3 ug 6 x 16.5 ug 6 x 100 ug 100 ug 300 ug 600 ug 100 ug 100 ug 600 ug 14.3 82.5 273.0 104.7 127.9 0 drug .14 .28 .46 1.1 1.3 Dose Splitting Factor 1 2 3.3 7.9 9.3 - 0 15 The splitting factor is defined as the product per unit drug following the protocol outlined in Example 15. In these divided by the single dose product per unit drug (PUD ) . For studies, varied doses of 1 ug , 5 ug , 10 ug , 25 ug , 50 ug , and example for treatment group 2 the value 0.28 or product values in between are used to determine dose response ( EPO ) per unit drug (mmRNA ) is divided by the single dose outcomes. Split dosing for a 100 ug total dose includes three product per unit drug of 0.14 . The result is 2. Likewise , for 20 or six doses of 1.6 ug , 4.2 ug , 8.3 ug , 16.6 ug , or values and treatment group 4 , the value 1.1 or product ( EPO ) per unit total doses equal to administration of the total dose selected . drug (mmRNA) is divided by the single dose product per unit drug of 0.14 . The result is 7.9 . Consequently , the dose Injection sites are chosen from the limbs or any body splitting factor ( DSF ) may be used as an indicator of the surface presenting enough area suitable for injection. This efficacy of a split dose regimen . For any single administra- 25 may also include a selection of injection depth to target the tion of a total daily dose, the DSF should be equal to 1 . Therefore any DSF greater than this value in a split dose regimen is an indication of increased efficacy. To determine the dose response trends, impact of injection site and impact of injection timing , studies are performed. In 30 these studies, varied doses of 1 ug , 5 ug , 10 ug , 25 ug , 50 ug , and values in between are used to determine dose response outcomes . Split dosing for a 100 ug total dose includes three or six doses of 1.6 ug , 4.2 ug , 8.3 ug , 16.6 ug , or values and total doses equal to administration of the total dose selected . 35 Injection sites are chosen from the limbs or any body dermis ( Intradermal), epidermis ( Epidermal ), subcutaneous tissue ( SC ) or muscle (IM) . Injection angle will vary based on targeted delivery site with injections targeting the intra dermal site to be 10-15 degree angles from the plane of the surface of the skin , between 20-45 degrees from the plane of the surface of the skin for subcutaneous injections and angles of between 60-90 degrees for injections substantially into the muscle. RNAIMAXTM surface presenting enough area suitable for injection. This Example 17. Routes of Administration may also include a selection of injection depth to target the dermis ( Intradermal ), epidermis (Epidermal ), subcutaneous tissue ( SC ) or muscle ( IM) . Injection angle will vary based Further studies were performed to investigate dosing on targeted delivery site with injections targeting the intra- 40 using different routes of administration. Following the pro dermal site to be 10-15 degree angles from the plane of the tocol outlined in Example 15 , 4 mice per group were dosed surface of the skin , between 20-45 degrees from the plane of the surface of the skin for subcutaneous injections and intramuscularly ( I.M. ) , intravenously ( IV) or subcutane angles of between 60-90 degrees for injections substantially into the muscle . Example 16 : Dose Response and Injection Site Selection and Timing To determine the dose response trends, impact of injection site and impact of injection timing, studies are performed ously ( S.C. ) by the dosing chart outlined in Table 8. Serum 45 was collected 13 hours post injection from all mice , tissue was collected from the site of injection from the intramus cular and subcutaneous group and the spleen , liver and kidneys were collected from the intravenous group . The results from the intramuscular group are show in FIG . 7A and the subcutaneous group results are shown in FIG . 7B . TABLE 8 Dosing Chart Group Treatment 1 2 3 4 5 6 Route Dose of mmRNA Total Dose Dosing Vehicle Lipoplex - human EPO mmRNA I.M. 4 x 100 ug + 30 % 4 x 70 ul Lipoplex Lipoplex - human EPO mmRNA I.M. 4 x 100 ug 4 x 70 ul Buffer Lipoplex Lipoplex -human EPO mmRNA S.C. 4 x 100 ug + 30 % 4 x 70 ul Lipoplex Lipoplex - human EPO mmRNA Lipoplex - human EPO mmRNA S.C. 4 x 100 ug 4 x 70 ul Buffer I.V. 200 ug + 30 % 140 ul Lipoplex Lipoplexed -Luciferase mmRNA I.M. 100 ug + 30 % 4 x 70 ul Lipoplex Lipoplex Lipoplex Lipoplex US 10,898,574 B2 74 73 TABLE 8 - continued Dosing Chart Group Treatment 7 8 9 10 11 Lipoplexed -Luciferase mmRNA Lipoplexed - Luciferase mmRNA Lipoplexed - Luciferase mmRNA Lipoplexed - human EPO mmRNA Formulation Buffer Total Dose Route Dose of mmRNA Dosing Vehicle I.M. 100 ug S.C. 100 ug + 30 % 4 x 70 ul Buffer S.C. 100 ug I.V. 200 ug + 30 % 4 x 70 ul Buffer Lipoplex Lipoplex I.M. 4x multi dosing Example 18 : In Vivo Delivery of Modified mRNA 4 x 70 ul Lipoplex 140 ul Lipoplex 4 x 70 ul Buffer 15 Modified RNA was delivered to C57 /BL6 mice intramus- cularly, subcutaneously, or intravenously to evaluate the bio - distribution of modified RNA using luciferase. A formulation buffer used with all delivery methods contained 150 mM sodium chloride, 2 mM calcium chloride, 2 mM Na + -phosphate which included 1.4 mM monobasic sodium phosphate and 0.6 mM of dibasic sodium phosphate , and 0.5 mM ethylenediaminetetraacetic acid (EDTA ) was adjusted using sodium hydroxide to reach a final pH of 6.5 before being filtered and sterilized . A 1x concentration was used as the delivery buffer. To create the lipoplexed solution deliv ered to the mice , in one vial 50 ug of RNA was equilibrated for 10 minutes at room temperature in the delivery buffer and in a second vial 10ul RNAiMAXTM was equilibrated for 10 minutes at room temperature in the delivery buffer. After equilibrium , the vials were combined and delivery buffer was added to reach a final volume of 100 ul which was then incubated for 20 minutes at room temperature . Luciferin was administered by intraperitoneal injection ( IP ) at 150 mg/kg to each mouse prior to imaging during the plateau phase of the luciferin exposure curve which was between 15 and 30 minutes . To create luciferin , 1 g of D - luciferin potassium or sodium salt was dissolved in 66.6 ml of distilled phosphate buffer solution (DPBS ) , not containing Mg2 + or Ca2 + , to make a 15 mg/ml solution . The solution was gently mixed and passed through a 0.2 um syringe filter, before being purged with nitrogen , aliquoted and frozen at -80 ° C. while being protected from light as much as possible . The solution was thawed using a waterbath if luciferin was not dissolved , gently mixed and kept on ice on the day of dosing. Whole body images were taken of each mouse 2 , 8 and 24 hours after dosing . Tissue images and serum was collected the left hind limb . The bioluminescence average for the luciferase expression signals for each group at 2 , 8 and 24 hours after dosing are shown in FIG . 8A for the left hind limb and FIG . 8B for the right hind limb . The biolumines 20 ug cence showed a positive signal at the injection site of the 5 and 50 ug modified RNA formulations containing and not containing lipoplex . B. Subcutaneous Administration Mice were subcutaneously ( S.C. ) administered either 25 modified luciferase mRNA (Naked -Luc ), lipoplexed modi fied luciferase mRNA (Lipoplex - luc ) , lipoplexed modified G - CSF mRNA ( Lipoplex - G - CSF ) or the formation buffer at a single dose of 50 ug of modified mRNA in an injection of 100 ul for each formulation. The bioluminescence 30 volume average for the luciferase expression signals for each group at 2 , 8 and 24 hours after dosing are shown in FIG . 8C . The bioluminescence showed a positive signal at the injection site of the 50 ug modified mRNA formulations containing 35 and not containing lipoplex . C. Intravenous Administration 40 45 from each mouse 24 hours after dosing. Mice administered doses intravenously had their liver, spleen , kidneys, lungs , 50 heart, peri -renal adipose tissue and thymus imaged . Mice administered doses intramuscularly or subcutaneously had their liver, spleen, kidneys , lungs, peri - renal adipose tissue , and muscle at the injection site . From the whole body images the bioluminescence was measured in photon per 55 second for each route of administration and dosing regimen . A. Intramuscular Administration Mice were intramuscularly ( I.M. ) administered either modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO : 10) (Naked -Luc ), lipoplexed modified lucifer- 60 ase mRNA (Lipoplex -luc ), lipoplexed modified granulocyte colony -stimulating factor (G - CSF) mRNA (mRNA sequence shown in SEQ ID NO : 4 ; poly -A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl) ( Lipoplex - Cytokine) or the formation buffer at a single dose 65 of 50 ug of modified RNA in an injection volume of 50 ul for each formulation in the right hind limb and a single dose of 5 ug of modified RNA in an injection volume of 50 ul in Mice were intravenously ( I.V.) administered either modi fied luciferase mRNA ( Naked -Luc ), lipoplexed modified luciferase mRNA (Lipoplex - luc ), lipoplexed modified G - CSF mRNA ( Lipoplex - G - CSF ) or the formation buffer at a single dose of 50 ug of modified mRNA in an injection volume of 100 ul for each formulation . The bioluminescence average for the luciferase expression signal in the spleen from each group at 2 hours after dosing is shown in FIG . 8D . The bioluminescence showed a positive signal in the spleen of the 50 ug modified mRNA formulations containing lipoplex . Example 19 : In Vivo Delivery Using Lipoplexes A. Human EPO Modified RNA Lipoplex A formulation containing 100 ug of modified human erythropoietin mRNA (mRNA sequence shown in SEQ ID NO : 7 ; poly -A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl) (EPO ; fully modified 5 -methylcytosine; N1-methylpseudouridine) was lipoplexed with 30 % by volume of RNAIMAXTM (Lipoplex -h - Epo - 46 ; Generation 2 or Gen2) in 50-70 uL delivered intramuscu larly to four C57 / BL6 mice . Other groups consisted of mice receiving an injection of the lipoplexed modified luciferase mRNA (Lipoplex - luc ) ( IVT cDNA sequence shown in SEQ ID NO : 10 ) which served as a control containing 100 ug of modified luciferase mRNA was lipoplexed with 30 % by volume of RNAIMAXTM or mice receiving an injection of the formulation buffer as negative control at a dose volume of 65 ul. 13 hours after the intramuscular injection, serum was collected from each mouse to measure the amount of US 10,898,574 B2 76 75 human EPO protein in the mouse serum by human EPO ul/eye . A formulation buffer is also administered by IVT to ELISA and the results are shown in FIG . 9 . a control group of rats in a dose volume of 5 ul/eye . Eyes B. Human G - CSF Modified RNA Lipoplex from treated rats can be collected after 18 hours post A formulation containing 100 ug of one of the two types injection for sectioning and lysating to determine whether of modified human G - CSF mRNA (mRNA sequence shown 5 mmRNA can be effectively delivered in vivo to the eye and in SEQ ID NO : 4 ; poly - A tail of approximately 160 nucleo- result in protein production, and to also determine the cell tides not shown in sequence ; 5 ' cap , Capl ) (G - CSF fully type( s) responsible for producing protein in vivo . modified with 5 -methylcytosine and pseudouridine ( G - CSF ) Intranasal Administration or G - CSF fully modified with 5 -methylcytosine and N1-methyl-pseudouridine ( G - CSF -N1) lipoplexed with 10 A formulation containing 100 ug of either modified 30 % by volume of RNAIMAXTM and delivered in 150 uL mCherry mRNA lipoplexed with 30 % by volume of intramuscularly (I.M) , in 150 L subcutaneously ( S.C ) and RNAIMAXTM , modified mCherry mRNA in saline, modi in 225 uL intravenously ( I.V) to C57 / BL6 mice . Three fied luciferase mRNA lipoplexed with 30% by volume of control groups were administered either 100 ug ofmodified RNAIMAXTM or modified luciferase mRNA in saline is luciferase mRNA ( IVT cDNA sequence shown in SEQ ID 15 delivered intranasally. A formulation buffer is also admin NO : 10) intramuscularly ( Luc -unsp I.M. ) or 150 ug of istered to a control group intranasally. Lungs may be col modified luciferase mRNA intravenously (Luc - unsp I.V.) or lected about 13 hours post instillation for sectioning ( for 150 uL of the formulation buffer intramuscularly (Buffer those receiving mCherry mRNA ) or homogenization ( for I.M. ) . 6 hours after administration of a formulation, serum those receiving luciferase mRNA ). These samples will be was collected from each mouse to measure the amount of 20 used to determine whether mmRNA can be effectively human G - CSF protein in the mouse serum by human G - CSF delivered in vivo to the lungs and result in protein produc ELISA and the results are shown in FIG . 10 . tion , and to also determine the cell type (s ) responsible for C. Human G - CSF Modified RNA Lipoplex Comparison producing protein in vivo . A formulation containing 100 ug of either modified human G - CSF mRNA lipoplexed with 30 % by volume of 25 Example 20 : In Vivo Delivery Using Varying Lipid RNAIMAXTM with a 5 -methylcytosine ( 5mc ) and a Ratios pseudouridine ( 4 ) modification (G - CSF - Gen1 - Lipoplex ) , modified human G - CSF mRNA with a 5mc and y modifi Modified mRNA was delivered to C57 /BL6 mice to cation in saline ( G - CSF - Gen1 - Saline) , modified human G- CSF mRNA with a N1-5 -methylcytosine (N1-5mc) and a 30 evaluate varying lipid ratios and the resulting protein expres W modification lipoplexed with 30% by volume of sion . Formulations of 100 ug modified human EPO mRNA RNAIMAXTM (G -CSF -Gen 2 -Lipoplex ), modified human (mRNA sequence shown in SEQ ID NO : 7 ; poly -A tail of G - CSF mRNA with a N1-5mc and y modification in saline approximately 160 nucleotides not shown in sequence ; 5 ' ( G - CSF -Gen2 -Saline ), modified luciferase with a 5mc and y cap , Capl ) lipoplexed with 10 % , 30 % or 50 % modification lipoplexed with 30 % by volume of 35 RNAIMAXTM , 100 ug modified luciferase mRNA ( IVT RNAIMAXTM (Luc -Lipoplex ), or modified luciferase cDNA sequence shown in SEQ ID NO : 10 ) lipoplexed with mRNA with a 5mc and y modification in saline (Luc -Saline ) 10% , 30 % or 50 % RNAIMAXTM or a formulation buffer was delivered intramuscularly ( I.M. ) or subcutaneously were administered intramuscularly to mice in a single 70 ul ( S.C. ) and a control group for each method of administration dose . Serum was collected 13 hours post injection to was giving a dose of 80 uL of the formulation buffer (F. 40 undergo a human EPO ELISA to determine the human EPO Buffer) to C57 / BL6 mice . 13 hours post injection serum and protein level in each mouse . The results of the human EPO tissue from the site of injection were collected from each ELISA , shown in FIG . 12 , show that modified human EPO mouse and analyzed by G - CSF ELISA to compare human G - CSF protein levels . The results of the human G - CSF expressed in the muscle is secreted into the serum for each of the different percentage of RNAIMAXTM . tion are shown in FIG . 11A , and the subcutaneous admin Example 21 : Intramuscular and Subcutaneous In Vivo Delivery in Mammals protein in mouse serum from the intramuscular administra- 45 istration results are shown in FIG . 11B . D. mCherry Modified RNA Lipoplex Comparison A formulation containing 100 ug of either modified 50 Modified human EPO mRNA (mRNA sequence shown in SEQ ID NO : 7 ; poly -A tail of approximately 160 nucleotides mCherry mRNA (mRNA sequence shown in SEQ ID NO : 5 ; not in sequence ; 5 ' cap , Capl ) formulated in saline poly -A tail of approximately 160 nucleotides not shown in was shown delivered to either C57 /BL6 mice or Sprague -Dawley sequence; 5 ' cap , Capl ) lipoplexed with 30% by volume of RNAIMAXTM or modified mCherry mRNA in saline is rats to evaluate the dose dependency on human EPO pro Intramuscular and Subcutaneous Administration delivered intramuscularly and subcutaneously to mice . A 55 modified duction. Rats were intramuscularly injected with 50 ul of the human EPO mRNA ( h -EPO ), modified luciferase formulation buffer is also administered to a control group of mice either intramuscularly or subcutaneously. The site of injection on the mice may be collected 17 hours post injection for sectioning to determine the cell type ( s) respon sible for producing protein . 60 mRNA ( Luc ) ( IVT DNA sequence shown in SEQ ID NO : 10) or the formulation buffer ( F.Buffer) as described in the dosing chart Table 9 . Mice were intramuscularly or subcutaneously injected with 50 ul of the modified human EPO mRNA ( h - EPO ), A formulation containing 10 ug of either modified modified luciferase mRNA ( Luc ) or the formulation buffer mCherry mRNA lipoplexed with RNAIMAXTM , modified ( F.Buffer) as described in the dosing chart Table 10. 13 hours mCherry mRNA in a formulation buffer, modified luciferase post injection blood was collected and serum was analyzed mRNA lipoplexed with RNAMAXTM , modified luciferase 65 to determine the amount human EPO for each mouse or rat. mRNA in a formulation buffer can be administered by The average and geometric mean in pg /ml for the rat study Intravitreal Administration intravitreal injection ( IVT ) in rats in a dose volume of 5 are also shown in Table 9 . US 10,898,574 B2 78 77 TABLE 9 Rat Study Group h - EPO h - EPO h - EPO h - EPO h - EPO Luc F. Buffer G G G G G G G Dose R # 2 R #1 R #3 R #4 18.5 14.7 21.2 mean R # 6 pg/ml pg /ml 74.2 67.7 161.5 79.4 23.9 101.5 150.3 46.3 34.1 28.7 66.9 85.4 31.2 R#5 150 ug 61.8 86.3 69.9 55.2 59 100 ug 69.4 77.8 48.2 17.6 101.9 50 ug 143.6 60.9 173.4 145.9 61.5 10 ug 7.8 11.8 30.9 36.2 40.6 46.2 18.1 1 ug 9.1 35.8 100 ug 34.1 36.5 13.5 13.7 #1 # 2 # 3 #4 #5 #6 #7 Geometric Avg. 24.5 18.7 20.3 67.1 25.4 22.4 18.5 15 Example 23. In Vitro Transfection of VEGF - A TABLE 10 Mouse Study Average Level in serum Route Treatment Group Dose pg/ml IM IM IM IM IM h- EPO h- EPO h - EPO h - EPO h- EPO Luc F. Buffer h - EPO 1 2 3 4 5 6 100 ug 96.2 63.5 18.7 25.9 2.6 1 2 3 100 ug 100 ug IM IM SC SC SC Luc F. Buffer 50 ug 25 ug 10 ug ug 100 ug 0 1.0 72.0 26.7 17.4 Human vascular endothelial growth factor - isoform A (VEGF- A ) modified mRNA (mRNA sequence shown in 20 SEQ ID NO : 12 ; poly -A tail of approximately 160 nucleo tides not shown in sequence ; 5 ' cap , Capl ) was transfected via reverse transfection in Human Keratinocyte cells in 24 multi -well plates. Human Keratinocytes cells were grown in EPILIFE® medium with Supplement S7 from Invitrogen 25 ( Carlsbad, Calif .) until they reached a confluence of 50-70% . The cells were transfected with 0 , 46.875 , 93.75 , 187.5 , 375 , 750 , and 1500 ng ofmodified mRNA (mmRNA ) encoding VEGF - A which had been complexed with RNAIMAXTM from Invitrogen ( Carlsbad, Calif . ). The RNA : 30 RNAIMAXTM complex was formed by first incubating the RNA with Supplement - free EPILIFE® media in a 5x volu metric dilution for 10 minutes at room temperature. In a Example 22 : Duration of Activity after Intramuscular In Vivo Delivery in Rats 35 Modified human EPO mRNA (mRNA sequence shown in SEQ ID NO : 7 ; poly -A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl ) formulated in saline was delivered to Sprague -Dawley rats to determine the duration of the dose response . Rats were intramuscularly injected with 50 ul of the modified human EPO mRNA (h- EPO ) , modified luciferase mRNA (IVT cDNA sequence shown in SEQ ID NO : 10 ) (Luc) or the formulation buffer ( F.Buffer) as described in the dosing chart Table 11. The rats were bled 2 , 6 , 12 , 24 , 48 and 72 hours after the intramus cular injection to determine the concentration of human EPO in serum at a given time . The average and geometric mean in pg /ml for this study are also shown in Table 11 . second vial , RNAIMAXTM reagent was incubated with Supplement- free EPILIFE® Media in a 10x volumetric dilution for 10 minutes at room temperature. The RNA vial was then mixed with the RNAIMAXTM vial and incubated for 20-30 minutes at room temperature before being added to the cells in a drop -wise fashion . The fully optimized mRNA encoding VEGF -A trans 40 fected with the Human Keratinocyte cells included modifi cations during translation such as natural nucleoside triphos phates (NTP ), pseudouridine at each uridine site and 5 -methylcytosine at each cytosine site (pseudo - U /5mC ), and 45 N1 -methyl-pseudouridine at each uridine site and 5 -meth ylcytosine at each cytosine site (N1-methyl- Pseudo - U / 5mC ) . Cells were transfected with the mmRNA encoding VEGF -A and secreted VEGF -A concentration ( pg /ml) in the culture medium was measured at 6 , 12 , 24 , and 48 hours post - transfection for each of the concentrations using an ELISA kit from Invitrogen ( Carlsbad , Calif.) following the TABLE 11 Dosing Chart Group Dose R #1 R #2 R #3 Luc 100 ug 60.0 62.4 53.6 100 ug 66.4 102.5 45.6 100 ug 132.9 55.1 89.0 100 g 51.1 76.3 264.3 100 ug 96.3 59.0 85.7 100 ug 46.3 66.9 73.5 24 , 48 and 72 hour 100 ug 60.2 38.5 48.8 F. Buffer 24 , 48 and 72 hour h - EPO h - EPO h - EPO h - EPO h - EPO h - EPO 2 6 12 24 48 72 hour hour hour hour hour hour 50.0 10.0 R #4 R# 5 R #6 R#7 33.2 68.6 66.4 72.8 78.1 56.8 122.5 8.1 80.1 85.6 105.6 63.3 142.4 77.6 73.5 75.0 82.6 63.5 80.3 69.7 57.3 136.7 110 46.1 3.6 26.1 80.9 54.7 Avg. pg Geometric ml mean pg /ml 59.6 68.6 87.4 108.6 77.9 58.2 55.8 84.5 95.3 77.0 80.1 37.2 48.9 29.2 75.8 10.4 US 10,898,574 B2 79 80 manufacturers recommended instructions. These data , shown in Table 12 , show that modified mRNA encoding VEGF -A is capable of being translated in Human Keratinocyte cells and that VEGF - A is transported out of the cells and bled at 13 hours after the intramuscular injection to deter mine the concentration of human the polypeptide in serum in pg /mL . The results revealed that administration of Factor released into the extracellular environment. lation buffer ( F.Buffer) at 2x100 ug /mouse. The mice were 5 IX mmRNA resulted in levels of 1600 pg mL / at 13 hours as TABLE 12 VEGF- A Dosing and Protein Secretion Dose (ng) 6 hours 12 hours 24 hours 48 hours (pg /ml) (pg /ml) (pg ml / ) (pg /ml) 10 VEGF- A Dose Containing Natural NTPs 46.875 93.75 187.5 375 750 1500 10.37 9.79 14.07 19.16 21.51 36.11 18.07 20.54 24.56 37.53 38.90 61.90 33.90 41.95 45.25 53.61 88.28 51.44 61.79 76.70 86.54 67.02 65.75 64.39 VEGF -A Dose Containing Pseudo - U / 5mC 46.875 93.75 187.5 375 750 1500 10.13 16.67 33.99 72.88 11.00 16.04 69.15 133.95 198.96 20.00 46.47 83.00 145.61 448.50 524.02 392.44 526.58 426.97 505.41 34.07 188.10 304.30 345.65 120.77 VEGF - A Dose Containing N1 -methyl- Pseudo - U /5mC 46.875 93.75 187.5 375 750 1500 0.03 12.37 104.55 605.89 445.41 261.61 6.02 46.38 365.71 1201.23 27.65 100.42 167.56 1056.91 121.23 1036.45 1025.41 1653.63 1522.86 714.68 1053.12 1889.23 1954.81 1513.39 Example 24. In Vivo Studies of Factor IX 15 compared to less than 100 pg/mL of Factor IX for either Luciferase or buffer control administration . Example 25. Multi -Site Administration : Intramuscular and Subcutaneous Human G - CSF mmRNA (mRNA sequence shown in SEQ ID NO : 4 ; poly - A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl ) modified as either Genl or Gen2 ( 5 -methylcytosine ( 5mc ) and a pseudouridine ( 4 ) modification , G - CSF - Genl ; or N1-5 -methylcytosine (N1 5mc ) and a y modification , G - CSF -Gen2 ) and formulated in saline were delivered to mice via intramuscular ( IM ) or 20 subcutaneous ( SC ) injection. Injection of four doses or 2x50 ug ( two sites ) daily for three days ( 24 hrs interval) was performed. The fourth dose was administered 6 hrs before blood collection and CBC analysis. Controls included Luciferase ( cDNA sequence for IVT shown in SEQ ID NO : 25 10) or the formulation buffer ( F.Buffer ). The mice were bled at 72 hours after the first mmRNA injection ( 6 hours after the last mmRNA dose ) to determine the effect of mmRNA encoded human G - CSF on the neutrophil count. The dosing regimen is shown in Table 13 as are the resulting neutrophil 30 counts (thousands/uL ). Asterisks indicate statistical signifi cance at p <0.05 . For intramuscular administration , the data reveal a four fold increase in neutrophil count above control at day 3 for 35 Human Factor IX mmRNA (mRNA shown in SEQ ID NO : 8 ; poly -A tail of approximately 160 nucleotides not the Gen1 G - CSF mmRNA and a two fold increase for the Gen2 G - CSF mmRNA . For subcutaneous administration, the data reveal a two fold increase in neutrophil count above control at day 3 for the Gen2 CT -CSF mmRNA TABLE 13 Dosing Regimen Dose Vol. Gr. Treatment 1 2 3 4 5 6 7 G- CSF ( Gen1 ) G- CSF ( Gen1 ) G- CSF ( Gen2 ) G- CSF ( Gen2 ) Luc (Gen1 ) Luc (Gen1) Luc ( Gen2 ) 8 Luc (Gen2) 9 F. Buffer 10 F. Buffer 11 Untreated shown in sequence ; 5 ' cap , Capl ) (Gen1; fully modified 5 -methylcytosine and pseudouridine ) formulated in saline was delivered to mice via intramuscular injection. The Route I.M S.C I.M S.C I.M. S.C. I.M S.C I.M S.C. Dosing N = Dose (ug /mouse ) (ul/mouse) Vehicle 5 2 x 50 ug ( four doses ) 50 F. buffer 5 2 x 50 ug ( four doses ) 50 F. buffer 5 2 x 50 ug ( four doses ) F. buffer 50 5 2 x 50 ug ( four doses ) 50 F. buffer 5 2 x 50 ug ( four doses ) 50 F. buffer 5 2 x 50 ug ( four doses ) 50 F. buffer 5 2 x 50 ug ( four doses ) 50 F. buffer 5 2 x 50 ug ( four doses ) 50 F. buffer 4 0 ( four doses ) 50 F. buffer 4 4 O ( four doses ) 50 F. buffer Neutrophil Thous/ uL 840 * 430 746 * 683 201 307 336 357 245 509 312 Example 26. Intravenous Administration Human G - CSF mmRNA (mRNA sequence shown in SEQ results demonstrate that Factor IX protein was elevated in 60 ID NO : 4 ; poly - A tail of approximately 160 nucleotides not serum as measured 13 hours after administration . shown in sequence ; 5 ' cap , Capl) modified with 5 -methyl In this study, mice (N= 5 for Factor IX , N=3 for Luciferase cytosine ( 5mc ) and a pseudouridine (W ) modification; or or Buffer controls) were intramuscularly injected with 50 ul having no modifications and formulated in 10 % lipoplex of the Factor IX mmRNA (mRNA sequence shown in SEQ (RNAIMAXTM ) were delivered to mice at a dose of 50 ug ID NO : 8 ; poly - A tail of approximately 160 nucleotides not 65 RNA and in a volume of 100 ul via intravenous (IV) shown in sequence ; 5 ' cap , Capl ) , Luciferase ( cDNA injection at days 0 , 2 and 4. Neutrophils were measured at sequence for IVT shown in SEQ ID NO : 10) or the formu- days 1 , 5 and 8. Controls included non -specific mammalian US 10,898,574 B2 81 82 RNA or the formulation buffer alone ( F.Buffer ). The mice were bled at days 1 , 5 and 8 to determine the effect of mmRNA - encoded human G - CSF to increase neutrophil count. The dosing regimen is shown in Table 14 as are the TABLE 15 resulting neutrophil counts ( thousands /uL ; K /uL ). Dosing Regimen 5 Dose For intravenous administration, the data reveal a four to five fold increase in neutrophil count above control at day 5 Group Treatment with G - CSF mmRNA but not with unmodified G - CSF G - CSF G - CSF mRNA or non - specific controls . Blood count returned to baseline four days after the final injection. No other changes in leukocyte populations were observed . An asterisk indicates statistical significance at p <0.001 compared to buffer. 10 G -CSF G - CSF EPO EPO EPO Vol. 1 2 3 G-CSF (Gen1) Day 1 G- CSF (Gen1 ) Day 5 G- CSF (Gen1) Day 8 4 G-CSF (no 5 5 5 5 Dosing (ul/mouse ) Vehicle 100 10% lipoplex 100 10% lipoplex 100 10% lipoplex 100 10% lipoplex Neutrophil 20 K /uL 2.91 5.32 * 25 2.06 1.88 modification ) Day 1 5 100 10% lipoplex 1.95 Day 5 G- CSF (no modification ) 5 100 10% lipoplex 2.09 RNA Control 5 100 10% lipoplex 2.90 5 100 1.68 4 100 4 100 4 100 4 100 10% lipoplex 10% lipoplex 10% lipoplex 10% lipoplex 10% lipoplex 5 G- CSF (no modification ) 6 Day 8 7 8 9 10 11 12 Day 1 RNA Control Day 5 RNA Control Day 8 F. Buffer Day 1 F. Buffer Day 5 F. Buffer Day 8 EPO Luciferase 50 50 50 50 50 5 5 5 5 5 Saline Saline F. buffer Saline Saline F. buffer F. buffer serum 19.8 0.5 0.5 191.5 15.0 4.8 Example 28. EPO Multi - Dose /Multi - Administration Dose N= Luciferase F. buffer Dosing (ul/mouse) Vehicle 15 TABLE 14 Dosing Regimen Gr. Treatment Vol. N= Average Protein Product pg /mL , 30 35 Control dosing involved use of buffer at a single dose . Human EPO blood levels were evaluated 13 hours post injection . 1.72 2.51 Studies utilizing multiple intramuscular injection sites at one time point were designed and performed . The design of a single multi -dose experiment involved using human erythropoietin ( EPO ) mmRNA (mRNA sequence shown in SEQ ID NO : 7 ; poly -A tail of approxi mately 160 nucleotides not shown in sequence ; 5 cap , Capl) or G - CSF (mRNA sequence shown in SEQ ID NO : 4 ; poly - A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl ) administered in saline . The dosing vehicle (F. buffer ) was used as a control. The EPO and G - CSF mmRNA were modified with 5 -methylcytosine at each cytosine and pseudouridine replacement at each uridine site . Animals ( n = 5 ) , Sprague- Dawley rats, were injected IM ( intramuscular) for the single unit dose of 100 ug ( delivered to one thigh ). For multi -dosing 6 doses of 100 ug ( delivered to two thighs ) were used for both EPO and G - CSF mmRNA . 40 1.31 Human EPO protein was measured in rat serum 13 hours post I.M. Five groups of rats were treated and evaluated . The results are shown in Table 16 . TABLE 16 1.92 Multi- dose study 45 Avg. Pg /mL Example 27. Saline Formulation : Intramuscular human Administration Human G - CSF mmRNA (mRNA sequence shown in SEQ ID NO : 4 ; poly - A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl) and human EPO mmRNA Group Treatment 50 (mRNA sequence shown in SEQ ID NO : 7 ; poly - A tail of approximately 160 nucleotides not shown in sequence; 5 ' 55 cap , Capl ) ; G - CSF mmRNA (modified with 5 -methylcyto sine ( 5mc ) and pseudouridine ( V )) and EPO mmRNA (modi fied with N1-5 -methylcytosine (N1-5mc ) and y modifica 1 2 3 4 5 Human EPO mmRNA Human EPO mmRNA G- CSF mmRNA G- CSF mmRNA Buffer Alone Dose of mmRNA 1 x 100 ug 6 x 100 mg 1 x 100 ug 6 x 100 ug Total Dose 100 ug 600 ug 100 ug 600 ug EPO , serum 143 256 43 58 20 Example 29. Signal Sequence Exchange Study tion ), were formulated in saline and delivered to mice via Several variants of mmRNAs encoding human Granulo intramuscular ( IM) injection at a dose of 100 ug . 60 cyte colony stimulating factor ( G -CSF) (mRNA sequence Controls included Luciferase ( IVT cDNA sequence shown in SEQ ID NO : 4 ; poly - A tail of approximately 160 shown in SEQ ID NO : 10 ) or the formulation buffer ( F.Buf- nucleotides not shown in sequence; 5 ' cap , Capl) were fer ). The mice were bled at 13 hours after the injection to synthesized using modified nucleotides pseudouridine and determine the concentration of the human polypeptide in 5 -methylcytosine (pseudo - U /5mC ). These variants included serum in pg /mL (G - CSF groups measured human G - CSF in 65 the G - CSF constructs encoding either the wild - type N mouse serum and EPO groups measured human EPO in terminal secretory signal peptide sequence mouse serum ). The data are shown in Table 15 . (MAGPATQSPMKLMALQLLLWHSALWTVQEA; SEQ US 10,898,574 B2 84 83 ID NO : 13 ) , no secretory signal peptide sequence , or secretory signal peptide sequences taken from other mRNAs. These included sequences where the wild type GCSF signal peptide sequence was replaced with the signal peptide of sequence either: human a - 1 -anti trypsin Transfection Preparation: 5 (MMPSSVSWGILLLAGLCCLVPVSLA ; SEQ ID NO : 14 ) , human Factor IX (MQRVNMIMAESPSLITI CLLGYLLSAECTVFLDHENANKILNRPKR ; SEQ ID NO : 15 ) , human Prolactin (MKGSLLLLL- VSNLLLCQSVAP; SEQ ID NO : 16 ) , or human Albumin ( MKWVTFISLLFLFSSAYSRGVFRR ; SEQ ID NO : 17 ) . 250 ng of modified mRNA encoding each G - CSF variant was transfected into HEK293A ( 293A in the table) , mouse myoblast ( MM in the table ) ( C2C12 , CRL - 1772, ATCC ) and rat myoblast ( RM in the table) ( L6 line , CRL - 1458 , ATCC ) mmRNA encoding human G - CSF (mRNA sequence shown in SEQ ID NO : 4 ; poly - A tail of approximately 160 nucleotides not shown in sequence ; 5 ' cap , Capl ) ( contain ing either ( 1 ) natural NTPs , ( 2 ) 100 % substitution with 5 -methyl cytidine and pseudouridine, or (3 ) 100 % substitu tion with 5 -methyl cytidine and N1 -methyl pseudouridine; 10 mmRNA encoding luciferase (IVT cDNA sequence shown in SEQ ID NO : 10 ) ( containing either ( 1 ) natural NTPs or (2 ) 100 % substitution with 5 -methyl cytidine and pseudou ridine) and TLR agonist R848 ( Invivogen tlrl - r848 ) were diluted to 38.4 ng/uL in a final volume of 2500 uL Optimem 15 1 . Separately, 432 uL of Lipofectamine 2000 (Invitrogen cell lines in a 24 well plate using 1 ul of Lipofectamine 2000 ( Life Technologies ), each well containing 300,000 cells . The 11668-027 , lot 1070962 ) was diluted with 13.1 mL supernatants were harvested after 24 hrs and the secreted Optimem I. In a 96 well plate nine aliquots of 135 uL of each G - CSF protein was analyzed by ELISA using the Human 20 mmRNA , positive control (R - 848 ) or negative control (Op G - CSF ELISA kit (Life Technologies ). The data shown in timem I ) was added to 135 uL of the diluted Lipofectamine Table 17 reveal that cells transfected with G - CSF mmRNA 2000. The plate containing the material to be transfected was encoding the Albumin signal peptide secrete at least 12 fold incubated for 20 minutes. The transfection mixtures were more G - CSF protein than its wild type counterpart. 25 then transferred to each of the human PBMC plates at 50 uL per well . The plates were then incubated at 37 C. At 2 , 4 , 8 , TABLE 17 20 , and 44 hours each plate was removed from the incubator, Signal Peptide Exchange and the supernatants were frozen . MM RM 293A After the last plate was removed, the supernatants were 30 Signal peptides (pg /ml) (pg/ml) ( pg /ml) G- CSF Natural a - 1 - anti trypsin 9650 9950 3450 6050 8475 Factor IX 11675 Prolactin Albumin 7875 No Signal peptide 122050 5000 6175 1525 81050 173300 0 0 0 assayed using a human G - CSF ELISA kit ( Invitrogen KHC2032 ) and human IFN - alpha ELISA kit ( Thermo Sci entific 41105-2 ) . Each condition was done in duplicate . 11675 9800 Example 30. Cytokine Study: PBMC PBMC Isolation and Culture : 50 mL of human blood from two donors was received Results : 35 The ability of unmodified and modified mRNA (mmR NAs) to produce the encoded protein was assessed (G - CSF production ) over time as was the ability of the mRNA to trigger innate immune recognition as measured by inter 40 feron - alpha production . Use of in vitro PBMC cultures is an accepted way to measure the immunostimulatory potential of oligonucleotides (Robbins et al . , Oligonucleotides 2009 from Research Blood Components (lots KP30928 and 19 : 89-102). KP30931 ) in sodium heparin tubes . For each donor, the blood was pooled and diluted to 70 mL with DPBS ( SAFC 45 Results were interpolated against the standard curve of Bioscience 59331C , lot 071M8408 ) and split evenly each ELISA plate using a four parameter logistic curve fit. between two 50 mL conical tubes . 10 mL of Ficoll Paque Shown in Tables 18 and 19 are the average from 2 separate ( GE Healthcare 17-5442-03 , lot 10074400 ) was gently dis- PBMC donors of the G - CSF and IFN - alpha production over pensed below the blood layer. The tubes were centrifuged at time as measured by specific ELISA . 2000 rpm for 30 minutes with low acceleration and braking . 50 In the G - CSF ELISA , background signal from the Lipo The tubes were removed and the buffy coat PMBC layers fectamine untreated condition was subtracted at each were gently transferred to a fresh 50 mL conical and washed timepoint. 2000 The data demonstrated specific production of with DPBS . The tubes were centrifuged at 1450 rpm for 10 human G - CSF protein by human peripheral blood mononu minutes . The supernatant was aspirated and the PBMC pellets were 55 clear is seen with G - CSF mRNA containing natural NTPs , resuspended and washed in 50 mL of DPBS . The tubes were 100 % substitution with 5 -methyl cytidine and pseudouri centrifuged at 1250 rpm for 10 minutes. This wash step was dine , or 100% substitution with 5 -methyl cytidine and repeated, and the PBMC pellets were resuspended in 19 mL N1-methyl pseudouridine. Production of G - CSF was sig of Optimem I (Gibco 11058, lot 1072088 ) and counted . The nificantly increased through the use of modified mRNA cell suspensions were adjusted to a concentration of 3.0x 60 relative to unmodified mRNA, with the 5 -methyl cytidine 10 ^ 6 cells /mL live cells . N1 - methyl pseudouridine containing G - CSF mmRNA These cells were then plated on five 96 well tissue culture and showing highest level of G - CSF production. With treated round bottom plates (Costar 3799 ) per donor at 50 ML regards totheinnate immune recognition, unmodified mRNA per well . Within 30 minutes, transfection mixtures were added to each well at a volume of 50 uL per well . After 4 65 resulted in substantial IFN - alpha production, while the hours post transfection , the media was supplemented with modified mRNA largely prevented interferon -alpha produc 10 uL of Fetal Bovine Serum ( Gibco 10082 , lot 1012368 ) tion . US 10,898,574 B2 85 86 TABLE 18 AMBION® , Austin , Tex .; MSCRIPTTM mRNA Production Kit , EPICENTRE® Biotechnologies, Madison , Wis . ). An in vitro transcription reaction contains between 1-2 ug of template DNA in the form of a linearized plasmid , PCR product, or single-stranded oligonucleotide with a double stranded polymerase promoter region . The template DNA encodes a strong translation initiation sequence such as a strong consensus Kozak sequence or an optimized , high G- CSF Signal G - CSF signal - 2 Donor Average pg /mL 2 Hr 4 Hr 20 Hr 44 Hr G - CSF ( 5mC /pseudouridine) G- CSF (5mC /N1-methyl pseudouridine ) 120.3 136.8 421.0 346.1 256.3 273.7 919.3 1603.3 431.8 1843.3 258.3 242.4 58.0 GCSF ( Natural-no modification ) Luciferase ( 5mC /pseudouridine) 8 Hr 63.5 92.6 129.6 4.5 153.7 33.0 186.5 TABLE 19 IFN- alpha signal 10 15 IFN -alpha signal - 2 donor average pg /mL 2 Hr 4 Hr 8 Hr G- CSF (5mC /pseudouridine) G- CSF ( 5mC /N1-methyl pseudouridine) 21.1 2.9 3.7 0.5 0.4 3.0 G- CSF (Natural) 0.0 0.4 39.1 0.8 2.1 0.4 151.3 17.2 23.3 Luciferase ( 5mC /pseudouridine) R - 848 Lipofectamine 2000 control 5 4.7 278.4 16.5 20 Hr 44 Hr 22.7 2.3 4.3 74.9 1.0 362.2 0.7 119.7 2.4 2.1 208.1 3.1 expression IRES including the EMCV IRES . Reaction vol umes are between 20-40 ul and contain 3 ' - O - Me - mP - G ( 5 ' ) ppp ( 5 ')G ARCA cap analog (NEW ENGLAND BIOLABS® ) in addition to an optimized ribonucleotide mixture of determined modified adenine, guanine, cytidine and uridine ribonucleotide analogs . Final reaction concen trations for nucleotide are 6 mM for the cap analog and 1.5-7.5 mM for each of the other nucleotides. The tempera ture and duration of the in vitro transcription reaction are optimized for efficiency, fidelity and yield. Reactions may be 20 incubated from 3-6 hours and up to 16 hours at 37 ° C. Following the in vitro transcription reaction, the capped mRNA undergoes polyadenylation using a commercially available poly -A tailing kit ( EPICENTRE® Biotechnolo gies , Madison , Wis . ). The resulting capped and polyade 25 nylated synthetic mRNA is then purified by denaturing agarose gel electrophoresis to confirm production of full length product and to remove any degradation products Example 31. Quantification in Exosomes followed by spin column filtration (RNeasy Kit , Qiagen , The quantity and localization of the mmRNA of the Valencia , Calif .; MEGACLEARTM AMBION® , Austin , present invention can be determined by measuring the 30 Tex . ). Purified synthetic mRNAs are resuspended in RNase amounts ( initial , timecourse , or residual basis ) in isolated free water containing an RNase inhibitor (RNASIN® Plus exosomes . In this study, since the mmRNA are typically RNase Inhibitor, Promega, Madison , Wis . ), quantified by codon - optimized and distinct in sequence from endogenous NANODROPTM ( Thermo Scientific , Logan, Utah ) and mRNA , the levels of mmRNA are quantitated as compared 35 stored at -20 ° C. to endogenous levels of native or wild type mRNA by using the methods of Gibbings , PCT/IB2009 /005878 , the contents Example 34 : Bulk Transfection of Modified mRNA into Cell Culture of which are incorporated herein by reference in their entirety. In these studies, the method is performed by first isolating A. Cationic Lipid Delivery Vehicles exosomes or vesicles preferably from a bodily fluid of a 40 RNA transfections are carried out using RNAIMax (Invit patient previously treated with a polynucleotide, primary rogen , Carlsbad , Calif .) or TRANSIT -mRNA (Mirus Bio , construct or mmRNA of the invention, then measuring, in Madison , Wis .) cationic lipid delivery vehicles. RNA and said exosomes , the polynucleotide, primary construct or reagent are first diluted in Opti - MEM basal media (Invitro mmRNA levels by one of mRNA microarray, qRT- PCR , or gen , Carlsbad, Calif. ). 100 ng/uL RNA is diluted 5x and 5 other means for measuring RNA in the art including by 45 uL of RNAIMax perm of RNA is diluted 10x . The diluted components are pooled and incubated 15 minutes at room suitable antibody or immunohistochemical methods. temperature before they are dispensed to culture media . For Example 32 : Bifunctional mmRNA TRANSIT -mRNA transfections , 100 ng /uL RNA is diluted 10x in Opti - MEM and BOOST reagent is added ( at a Using the teachings and synthesis methods described 50 concentration of 2 ul perm of RNA ), TRANSIT -mRNA is herein , modified RNAs are designed and synthesized to be added ( at a concentration of 2 uL perm of RNA ), and then bifunctional, thereby encoding one or more cytotoxic pro- the RNA - lipid complexes are delivered to the culture media tein molecules as well as be synthesized using cytotoxic after a 2 -minute incubation at room temperature. RNA nucleosides. transfections are performed in Nutristem xenofree hES Administration of the bifunctional modified mRNAs is 55 media (STEMGENT® , Cambridge, Mass . ) for RiPS deri effected using either saline or a lipid carrier. Once admin- vations , Dermal Cell Basal Medium plus Keratinocyte istered , the bifunctional modified mRNA is translated to Growth Kit ( ATCC ) for keratinocyte experiments, and Opti produce the encoded cytotoxic peptide. Upon degradation of MEM plus 2 % FBS for all other experiments. Successful the delivered modified mRNA , the cytotoxic nucleosides are introduction of a modified mRNA (mmRNA ) into host cells released which also effect therapeutic benefit to the subject. 60 can be monitored using various known methods, such as a fluorescent marker, such as Green Fluorescent Protein Example 33. Synthesis of Modified mRNA (GFP ) . Successful transfection of a modified mRNA can also be determined by measuring the protein expression Modified mRNA is generated from a cDNA template level of the target polypeptide by e.g. , Western Blotting or containing a 17 RNA -polymerase promoter sequence using 65 immunocytochemistry. Similar methods may be followed a commercially available 17 RNA polymerase transcription for large volume scale -up to multi - liter ( 5-10,000 L ) culture kit (MEGASCRIPT® High Yield Transcription KIT, format following similar RNA - lipid complex ratios. US 10,898,574 B2 87 mRNA Transcripts 88 trolled via rotameters. Air was sparged into the reactor via a submerged frit ( 5 um pore size ) and through the reactor head cally detect the exogenous transcripts. Discharging a 150 uF capacitor charged to F into 2.5x10 cells suspended in 50 ul of Opti - MEM ( Invitrogen , Carlsbad , Calif.) in a standard used for cell counting was stained with trypan blue ( Sigma , St. Louis , Mo. ) . Cell count and cell viability determination were performed via hemocytometry using a microscope. For repeated delivery in excess of 10,000 copies of modified mRNA transcripts per cell , as determined using the standard curve method, while maintaining high viability (> 70 % ). Further experiments may reveal that the voltage required to efficiently transfect cells with mmRNA transcripts can 15 depend on the cell density during electroporation . Cell density may vary from 1x10 cell / 50 ul to a density of 2.5x10 cells / 50 ul and require from 110V to 145V to transfect cells with similar efficiencies measured in transcript copies per cell . Large multi- liter ( 5-10,000 L ) elec- 20 troporation may be performed similar to large volume flow electroporation strategies similar to methods described with for 20 minutes at 2000 rpm ( 4 ° C. ) for cell separation. Supernatant was analyzed for the following parameters: titer, sialic acid , glucose , lactate , glutamine, glutamate, pH , p02 , pCO2 , ammonia , and, optionally, lactate dehydroge nase (LDH ). Additional back-up samples were frozen at -20 ° C. To measure secreted humanized IgG antibody titers , supernatant is taken from seed - stock cultures of all stable cell pools , the IgG titer is determined by ELISA and divided by the mean number of cells to calculate the specific productivity. The highest values are the cell pools with the Ser132A CERT mutant, followed by wild type CERT. In both, IgG expression is markedly enhanced compared to B. Electroporation Delivery of Exogenous Synthetic Electroporation parameters are optimized by transfecting space for CO2 removal. Molecular oxygen was sparged MRC - 5 fibroblasts with in vitro synthetic modified mRNA through the same frit for DO control. CO2 was sparged ( mmRNA ) transcripts and measuring transfection efficiency 5 through same frit as used for pH control. Samples of cells by quantitative RT - PCR with primers designed to specifi- were removed from the reactor on a daily basis . A sample electroporation cuvette with a 2 mm gap is sufficient for 10 analysis of metabolites, additional samples were centrifuged the above described constraints (Li et al . , 200 Geng et al . , carrier -alone or untransfected cells . Continuous or Batch -Fed Culture 2010 ) . 25 Example 35. Overexpression of Ceramide Transfer Protein to Increase Therapeutic Antibody Protein Production in Established CHO Cell Lines A. Batch Culture 30 An antibody producing CHO cell line (CHO DG44 ) secreting a humanized therapeutic IgG antibody is transfected a single time with lipid cationic delivery agent alone ( control) or a synthetic mRNA transcript encoding wild type ceramide transfer protein ( CERT) or a non -phosphorylation 35 competent Ser132A CERT mutant. The sequences are taught in for example, U.S. Ser. No. 13 /252,049 , the contents of which are incorporated herein by reference in their entirety. CERT is an essential cytosolic protein in mammalian cells that transfers the sphingolipid ceramide from the endoplas- 40 mic reticulum to the Golgi complex where it is converted to sphingomyelin ( Hanada et al . , 2003 ). Overexpression of CERT significantly enhances the transport of secreted proteins to the plasma membrane and improves the production of proteins that are transported via the secretory pathway 45 from eukaryotic cells thereby enhancing secretion of proteins in the culture medium . Synthetic mRNA transcripts are pre -mixed with a lipid cationic delivery agent at a 2-5 : 1 carrier :RNA ratio . The initial seeding density is about 2x105 viable cells /mL . The synthetic mRNA transcript is delivered 50 after initial culture seeding during the exponential culture growth phase to achieve a final synthetic mRNA copy number between 10x102 and 10x109 per cell . The basal cell culture medium used for all phases of cell inoculum generation and for growth of cultures in bioreactors was modi- 55 fied CD - CHO medium containing glutamine , sodium bicarbonate , insulin and methotrexate . The pH of the medium was adjusted to 7.0 with 1 N HCl or IN NaOH after addition of all components. Culture run times ended on days 7 , 14 , 21 or 28+ . Production - level 50 L scale reactors ( stainless steel 60 reactor with two marine impellers ) were used and are scalable to > 10,000 L stainless steel reactors ( described in commonly - assigned patent application U.S. Ser. No. 60/436 , 050 , filed Dec. 23 , 2002 , and U.S. Ser. No. 10/740,645 ) . A data acquisition system ( Intellution Fix 32 , OSIsoft, LLC , 65 San Leandro , Calif.) recorded temperature, pH , and dissolved oxygen (DO ) throughout runs. Gas flows were con- An antibody producing CHO cell line (CHO DG44 ) secreting humanized IgG antibody is transfected with lipid cationic delivery agent alone ( control) or a synthetic mRNA transcript encoding wild type ceramide transfer protein or a non -phosphorylation competent Ser132A CERT mutant. Synthetic mRNA transcripts are pre -mixed with a lipid cationic delivery agent at a 2-5 : 1 carrier :RNA ratio . The initial seeding density was about 2x10% viable cells/mL . Synthetic mRNA transcript is delivered after initial culture seeding during the exponential culture growth phase to achieve a final synthetic mRNA copy number between 10x102 and 10x103 per cell . The basal cell culture medium used for all phases of cell inoculum generation and for growth of cultures in bioreactors was modified CD - CHO medium containing glutamine, sodium bicarbonate , insulin and methotrexate . The pH of the medium was adjusted to 7.0 with 1 N HCl or IN NaOH after addition of all components . Bioreactors of 5 L scale (glass reactor with one marine impeller ) were used to obtain maximum CERT protein production and secreted humanized IgG antibody curves . For continuous or fed - batch cultures, the culturing run time is increased by supplementing the culture medium one or more times daily (or continuously ) with fresh medium during the run . In the a continuous and fed -batch feeding regimens, the cultures receive feeding medium as a continu ously - supplied infusion , or other automated addition to the culture, in a timed , regulated, and / or programmed fashion so as to achieve and maintain the appropriate amount of synthetic mRNA : carrier in the culture. The preferred method is a feeding regimen of a once per day bolus feed with feeding medium containing synthetic mRNA : carrier on each day of the culture run , from the beginning of the culture run to the day of harvesting the cells . The daily feed amount was recorded on batch sheets. Production - level 50 L scale reactors ( stainless steel reactor with two marine impellers ) were used and are scalable to > 10,000 L stainless steel reactors. A data acquisition system ( Intellution Fix 32 ) recorded temperature , pH , and dissolved oxygen ( DO ) throughout runs. Gas flows were controlled via rotameters. Air was sparged into the reactor via a submerged frit ( 5 um pore size) and through the reactor head space for CO2 removal. Molecular oxygen was sparged through the same frit for DO control. CO2 was sparged through same frit as US 10,898,574 B2 89 90 used for pH control. Samples of cells were removed from the reactor on a daily basis . A sample used for cell counting was stained with trypan blue ( Sigma , St. Louis , Mo. ) . Cell count and cell viability determination were performed via hemo- B. Continuous or Batch - Fed Culture A primary CHO cell line derived and expanded as described above ( see Example 36a ) is transfected with lipid cationic delivery agent alone ( control) or a synthetic mRNA cytometry using a microscope. For analysis of metabolites, 5 transcript encoding human erythropoietin protein . Synthetic additional samples were centrifuged for 20 minutes at 2000 mRNA transcripts are pre -mixed with a lipid cationic deliv rpm ( 4 ° C. ) for cell separation. Supernatant was analyzed for ery agent at a 2-5 : 1 carrier :RNA ratio . The initial seeding the following parameters: titer, sialic acid , glucose , lactate , density was about 2x10 % viable cells/mL . Synthetic mRNA glutamine , glutamate , pH , pO2 , pCO2 , ammonia, and, transcript is delivered after initial culture seeding during the optionally, lactate dehydrogenase (LDH ). Additional back- 10 exponential culture growth phase to achieve a final synthetic up samples were frozen at -20 ° C. To measure secreted mRNA copy number between 10x102 and 10x103 per cell . humanized IgG antibody titers, supernatant is taken from Culture conditions were as described above ( Example 35a) . seed - stock cultures of all stable cell pools , the IgG titer is For continuous or fed -batch cultures, the culturing run time determined by ELISA and divided by the mean number of is increased by supplementing the culture medium one or cells to calculate the specific productivity. The highest 15 more times daily (or continuously ) with fresh medium values are the cell pools with the Ser132A CERT mutant, during the run . In the a continuous and fed -batch feeding followed by wild type CERT. In both , IgG expression is regimens, the cultures receive feeding medium as a continu markedly enhanced compared to carrier - alone or untrans- ously - supplied infusion , or other automated addition to the culture, in a timed , regulated, and / or programmed fashion so fected cells . 20 as to achieve and maintain the appropriate amount of Example 36. De Novo Generation of a Mammalian synthetic mRNA : carrier in the culture. The preferred Cell Line Expressing Human Erythropoietin as a method is a feeding regimen of a once per day bolus feed Therapeutic Agent with feeding medium containing synthetic mRNA : carrier on each day of the culture run , from the beginning of the culture A. Batch Culture 25 run to the day of harvesting the cells . The daily feed amount This Example describes the production of human eryth- was recorded on batch sheets. Production - level 50 L scale ropoietin protein ( EPO ) from cultured primary CHO cells . reactors ( stainless steel reactor with two marine impellers ) Erythropoietin is a glycoprotein hormone that is required for were used and are scalable to > 10,000 L stainless steel red blood cell synthesis. EPO protein may be used as a reactors. Culture growth and analysis were performed as therapeutic agent for anemia from cancer, heart failure, 30 described herein ( see Example 35 ) . chronic kidney disease and myelodysplasia . Primary CHO It is to be understood that the words which have been used cells are isolated and cultured as described ( Tjio and Puck , are words of description rather than limitation , and that 1958 ) . Primary CHO cells were then expanded in modified changes may be made within the purview of the appended CD - CHO medium containing glutamine , sodium bicarbon- claims without departing from the true scope and spirit of the ate , insulin, and methotrexate ( see Example 35 ) using T -75 35 invention in its broader aspects . flasks ( Corning, Corning, N.Y.) and 250 and 500 mL spinWhile the present invention has been described at some ners (Bellco , Vineland , N.J. ) . T - flasks and spinners were length and with some particularity with respect to the several incubated at 37 ° C. in 6 % CO2 . After sufficient inoculum described embodiments, it is not intended that it should be was generated, the culture was transferred into a either a 5 limited to any such particulars or embodiments or any Lor a 50 L bioreactor as described above ( see Example 35 ) . 40 particular embodiment, but it is to be construed with refer Synthetic mRNA transcript encoding the human erythropoi- ences to the appended claims so as to provide the broadest etin protein are pre -mixed with a lipid cationic delivery possible interpretation of such claims in view of the prior art agent at a 2-5 : 1 carrier:RNA in a minimum of 1 % total and , therefore, to effectively encompass the intended scope culture volume . The initial seeding density is about 2x105 of the invention . viable cells /mL . The synthetic mRNA transcript is delivered 45 All publications , patent applications, patents, and other after initial culture seeding during the exponential culture references mentioned herein are incorporated by reference in growth phase to achieve a final synthetic mRNA copy their entirety. In case of conflict, the present specification, number between 10x102 and 10x103 per cell . Culture including definitions, will control. In addition , section head growth and analysis were performed as described above ( see ings , the materials, methods, and examples are illustrative Example 34 ) . only and not intended to be limiting . SEQUENCE LISTING < 160 > NUMBER OF SEQ ID NOS : 17 < 210 > SEQ ID NO 1 < 211 > LENGTH : 624 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 1 atggctggac ctgccaccca gagccccatg aagctgatgg ccctgcagct gctgctgtgg 60 cacagtgcac tctggacagt gcaggaagcc acccccctgg gccctgccag ctccctgccc 120 cagagcttcc tgctcaagtg cttagagcaa gtgaggaaga tccagggcga tggcgcagcg 180 US 10,898,574 B2 92 91 - continued ctccaggaga agctggtgag tgagtgtgcc acctacaagc tgtgccaccc cgaggagctg 240 gtgctgctcg gacactctct gggcatcccc tgggctcccc tgagcagctg ccccagccag 300 gccctgcagc tggcaggctg cttgagccaa ctccatagcg gccttttcct ctaccagggg 360 ctcctgcagg ccctggaagg gatctccccc gagttgggtc ccaccttgga cacactgcag 420 ctggacgtcg ccgactttgc caccaccato tggcagcaga tggaagaact gggaatggcc 480 cctgccctgc agcccaccca gggtgccatg ccggccttcg cctctgcttt ccagcgccgg 540 gcaggagggg tcctggttgc ctcccatctg cagagcttcc tggaggtgtc gtaccgcgtt 600 ctacgccacc ttgcccagcc ctga 624 < 210 > SEQ ID NO 2 < 211 > LENGTH : 829 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 2 ttggaccctc gtacagaagc taatacgact cactataggg aaataagaga gaaaagaaga gtaagaagaa atataagagc caccatggct ggacctgcca cccagagccc catgaagctg 120 atggccctgc agctgctgct gtggcacagt gcactctgga cagtgcagga agccaccccc 180 ctgggccctg ccagctccct gccccagagc ttcctgctca agtgcttaga gcaagtgagg 240 aagatccagg gcgatggcgc agcgctccag gagaagctgg tgagtgagtg tgccacctac aagctgtgcc accccgagga gctggtgctg ctcggacact ctctgggcat cccctgggct 300 cccctgagca gctgccccag ccaggccctg cagctggcag gctgcttgag ccaactccat 420 agcggccttt tcctctacca ggggctcctg caggccctgg aagggatctc ccccgagttg 480 ggtcccacct tggacacact gcagctggac gtcgccgact ttgccaccac catctggcag 540 cagatggaag aactgggaat ggcccctgcc ctgcagcoca cccagggtgc catgccggcc 600 ttcgcctctg ctttccagcg ccgggcagga ggggtcctgg ttgcctccca tctgcagage 660 ttcctggagg tgtcgtaccg cgttctacgc caccttgccc agccctgaag cgctgccttc 720 tgcggggctt gccttctggc catgcccttc ttctctccct tgcacctgta cctcttggtc tttgaataaa gcctgagtag gaaggcggcc gctcgagcat gcatctaga 780 60 360 829 < 210 > SEQ ID NO 3 < 211 > LENGTH : 772 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 3 ttggaccctc gtacagaagc taatacgact cactataggg aaataagaga gaaaagaaga 60 gtaagaagaa atataagagc caccatggcc ctgcagttgc tgctttggca ctcggccctc 120 tggacagtcc aagaagcgac tcctctcgga cctgcctcat cgttgccgca gtcattcctt 180 ttgaagtgtc tggagcaggt gcgaaagatt cagggcgatg gagccgcact ccaagagaag 240 ctctgcgcga catacaaact ttgccatccc gaggagctcg tactgctcgg gcacagcttg gggattccct gggctcctct ctcgtcctgt ccgtcgcagg ctttgcagtt ggcagggtgc 300 ctttcccagc tccactccgg tttgttcttg tatcagggac tgctgcaagc ccttgaggga 420 CCC gacgctggac ac -tgcagc tcgacg ggc ggatttcgca 480 acaaccatct ggcagcagat ggaggaactg gggatggcac ccgcgctgca gcccacgcag 540 ggggcaatgc cggcctttgc gtccgcgttt cagcgcaggg cgggtggagt cctcgtagcg 600 ata cag aat 360 US 10,898,574 B2 94 93 - continued agccaccttc aatcattttt ggaagtctcg taccgggtgc tgagacatct tgcgcagccg 660 tgagccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 720 ctcttggtct ttgaataaag cctgagtagg aaggcggccg ctcgagcatg ca 772 < 210 > SEQ ID NO 4 < 211 > LENGTH : 746 < 212 > TYPE : RNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 4 cucacuauag ggaaauaaga gagaaaagaa gaguaagaag aaauauaaga gccaccaaug 60 goccugcagu ugcugcuuug gcacucggco cucuggacag uccaagaagc gacuccucuc 120 ggaccugccu caucguugcc gcagucauuc cuuuugaagu gucuggagca ggugcgaaag 180 auucagggcg auggagccgc acuccaagag aagcucugcg cgacauacaa acuuugccau 240 cccgaggagc ucguacugcu cgggcacagc uuggggauuc ccugggcucc ucucucgucc 300 uguccgucgc aggcuuugca guuggcaggg ugccuuuccc agcuccacuc cgguuuguuc uuguaucagg gacugcugca agcccuugag ggaaucucgc cagaauuggg cccgacgcug 360 gacacguugc agcucgacgu ggcggauuuc gcaacaacca ucuggcagca gauggaggaa 480 cuggggaugg cacccgcgcu gcagcccacg cagggggcaa ugccggccuu ugcguccgcg 540 uuucagcgca gggcgggugg aguccucgua gcgagccacc uucaaucauu uuuggaaguc 600 ucguaccggg ugcugagaca ucuugcgcag ccgugagccu ucugcggggc uugccuucug 660 gccaugcccu ucuucucucc cuugcaccug uaccucuugg ucuuugaaua aagccugagu 720 aggaaggcgg ccgcucgagc augcau 746 420 < 210 > SEQ ID NO 5 < 211 > LENGTH : 854 < 212 > TYPE : RNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 5 gggaaauaag agagaaaaga agaguaagaa gaaauauaag agccaccaug guauccaagg 60 gggaggagga caacauggcg aucaucaagg aguucaugcg auucaaggug cacauggaag guucggucaa cggacacgaa uuugaaaucg aaggagaggg ugaaggaagg cccuaugaag ggacacagac cgcgaaacuc aaggucacga aagggggacc acuuccuuuc gccugggaca 120 240 uucuuucgcc ccaguuuaug uacgggucca aagcauaugu gaagcauccc gccgauauuc 300 cugacuaucu gaaacucagc uuucccgagg gauucaagug ggagcggguc augaacuuug 360 aggacggggg uguagucacc guaacccaag acucaagccu ccaagacggc gaguucaucu 420 acaaggucaa acugcggggg acuaacuuuc cgucggaugg gccggugaug cagaagaaaa 480 cgaugggaug ggaagcguca ucggagagga uguacccaga agauggugca uugaaggggg 540 agaucaagca gagacugaag uugaaagaug ggggacauua ugaugccgag gugaaaacga 600 cauacaaagc gaaaaagccg gugcagcuuc ccggagcgua uaaugugaau aucaaguugg 660 auauuacuuc acacaaugag gacuacacaa uugucgaaca guacgaacgc gcugagggua 720 gacacucgac gggaggcaug gacgaguugu acaaaugaua agcugccuuc ugcggggcuu 780 gccuucuggc caugcccuuc uucucucccu ugcaccugua ccucuugguc uuugaauaaa 840 gccugaguag gaag 854 180 US 10,898,574 B2 95 96 - continued < 210 > SEQ ID NO 6 < 211 > LENGTH : 924 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 6 tcaagctttt ggaccctcgt acagaagcta atacgactca ctatagggaa ataagagaga 60 aaagaagagt aagaagaaat ataagagcca ccatggtatc caagggggag gaggacaaca 120 tggcgatcat caaggagttc atgcgattca aggtgcacat ggaaggttcg gtcaacggac 180 acgaatttga aatcgaagga gagggtgaag gaaggcccta tgaagggaca cagaccgcga aactcaaggt cacgaaaggg ggaccacttc ctttcgcctg ggacattctt tcgccccagt 240 ttatgtacgg gtccaaagca tatgtgaagc atcccgccga tattcctgac tatctgaaac 360 tcagctttcc cgagggattc aagtgggagc gggtcatgaa ctttgaggac gggggtgtag tcaccgtaac ccaagactca agcctccaag acggcgagtt catctacaag gtcaaactgc 420 gggggactaa ctttccgtcg gatgggccgg tgatgcagaa gaaaacgatg ggatgggaag 540 cgtcatcgga gaggatgtac ccagaagatg gtgcattgaa gggggagatc aagcagagac 600 tgaagttgaa agatggggga cattatgatg ccgaggtgaa aacgacatac aaagcgaaaa 660 agccggtgca gcttcccgga gcgtataatg tgaatatcaa gttggatatt acttcacaca atgaggacta cacaattgtc gaacagtacg aacgcgctga gggtagacac tcgacgggag gcatggacga gttgtacaaa tgataagctg ccttctgcgg ggcttgcctt ctggccatgo 720 780 ccttcttctc tcccttgcac ctgtacctct tggtctttga ataaagcctg agtaggaagg 900 cggccgctcg agcatgcatc taga 924 300 480 840 < 210 > SEQ ID NO 7 < 211 > LENGTH : 725 < 212 > TYPE : RNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 7 gggaaauaag agagaaaaga agaguaagaa gaaauauaag agccaccaug ggagugcacg 60 agugucccgc gugguugugg uugcugcugu cgcucuugag ccucccacug ggacugccug ugcugggggc accacccaga uugaucugcg acucacgggu acuugagagg uaccuucuug 120 180 aagccaaaga agccgaaaac aucacaaccg gaugcgccga gcacugcucc cucaaugaga 240 acauuacugu accggauaca aaggucaauu ucuaugcaug gaagagaaug gaaguaggac 300 agcaggccgu cgaagugugg caggggcucg cgcuuuuguc ggaggcggug uugcgggguc 360 aggcccuccu cgucaacuca ucacagccgu gggagccccu ccaacuucau gucgauaaag 420 cggugucggg gcuccgcagc uugacgacgu ugcuucgggc ucugggcgca caaaaggagg 480 cuauuucgcc gccugacgcg gccuccgcgg caccccuccg aacgaucacc gcggacacgu 540 uuaggaagcu uuuuagagug uacagcaauu uccuccgcgg aaagcugaaa uuguauacug 600 gugaagcgug uaggacaggg gaucgcugau aagcugccuu cugcggggcu ugccuucugg 660 ccaugcccuu cuucucuccc uugcaccugu accucuuggu cuuugaauaa agccugagua 720 ggaag 725 < 210 > SEQ ID NO 8 < 211 > LENGTH : 1536 < 212 > TYPE : RNA < 213 > ORGANISM : Homo sapiens US 10,898,574 B2 98 97 - continued < 400 > SEQUENCE : 8 gggaaauaag agagaaaaga agaguaagaa gaaauauaag agccaccaau gcagcgcguc 60 aacaugauua uggccgaauc gccgggacuc aucacaaucu gccucuuggg uuaucucuug 120 ucggcagaau guaccguguu cuuggaucac gaaaacgcga acaaaauucu uaaucgcccg 180 aagcgguaua acuccgggaa acuugaggag uuugugcagg gcaaucuuga acgagagugc 240 auggaggaga aaugcuccuu ugaggaggcg agggaagugu uugaaaacacagagcgaaca 300 acggaguuuu ggaagcaaua cguag?uggg gaccagugug agucgaaucc gugccucaau 360 gggggaucau guaaagauga caucaauagc uaugaaugcu ggugcccguu uggguuugaa 420 gggaagaacu gugagcugga ugugacgugc aacaucaaaa acggacgcug ugagcaguuu 480 uguaagaacu cggcugacaa uaagguagua ugcucgugca cagagggaua ccggcuggcg 540 gagaaccaaa aaucgugcga gcccgcaguc ccguucccuu gugggagggu gagcguguca 600 cagacuagca aguugacgag agcggagacu guauuccccg acguggacua cgucaacagc accgaagccg aaacaaucou cgauaacauc acgcagagca cucaguccuu caaugacuuu 660 acgagggucg uaggugguga ggacgcgaaa cccggucagu uccccuggca ggugguauug 780 aacggaaaag ucgaugccuu uuguggaggu uccauuguca acgagaagug gauugucaca 840 gcggcacacu gcguagaaac aggagugaaa aucacgguag uggcgggaga gcauaacauu 900 gaagagacag agcacacgga acaaaagcga aaugucauca gaaucauucc acaccauaac uauaacgcgg caaucaauaa guacaaucac gacaucgcac uuuuggagcu ugacgaaccu 960 1020 uuggugcuua auucguacgu caccccuauu uguauugccg acaaagagua uacaaacauc 1080 uucuugaaau ucggcuccgg guacguaucg ggcuggggca gaguguucca uaaggguaga uccgcacugg uguugcaaua ccucagggug ccccucgugg aucgagecac uugucugcgg 1140 1200 uccaccaaau ucacaaucua caacaauaug uucugugcgg gauuccauga aggugggaga 1260 gauagcugcc agggagacuc aggggguccc cacgugacgg aagucgaggg gacgucauuu 1320 cugacgggaa uuaucucaug gggagaggaa ugugcgauga aggggaaaua uggcaucuac 1380 acuaaagugu cacgguaugu caauuggauc aaggaaaaga cgaaacucac gugaucagcc agcgcugccu ucugcggggc uugccuucug gccaugcccu ucuucucucc cuugcaccug 1440 uaccucuugg ucuuugaaua aagccugagu aggaag 1536 720 1500 < 210 > SEQ ID NO 9 < 211 > LENGTH : 794 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 9 caagcttttg gaccctcgta cagaagctaa tacgactcac tatagggaaa taagagagaa 60 aagaagagta agaagaaata taagagecac catgggagtg cacgagtgtc ccgcgtggtt 120 gtggttgctgctgtcgctct tgagcctccc actgggactg cctgtgctgg gggcaccacc 180 cagattgate tgcgactcac gggtacttga gaggtacctt cttgaagcca aagaagccga 240 aaacatcaca accggatgcgccgagcactg ctccctcaat gagaacatta ctgtaccgga tacaaaggtc aatttctatg catggaagag aatggaagta ggacagcagg ccgtcgaagt 300 gtggcagggg ctcgcgcttt tgtcggaggc ggtgttgcgg ggtcaggccc tcctcgtcaa 420 ctcatcacag ccgtgggagc ccctccaact tcatgtcgat aaagcggtgt cggggctccg 480 360 US 10,898,574 B2 99 100 - continued cagettgacg acgttgcttc gggctctggg cgcacaaaag gaggotattt cgccgcctga 540 cgcggcctcc gcggcacccc tccgaacgat caccgcggac acgtttagga agctttttag 600 agtgtacagc aatttcctcc gcggaaagct gaaattgtat actggtgaag cgtgtaggac aggggatcgc tgataagctg ccttctgcgg ggcttgcctt ctggccatgc ccttcttctc 660 tcccttgcac ctgtacctct tggtctttga ataaagcctg agtaggaagg cggccgctcg 780 agcatgcatc taga 794 720 < 210 > SEQ ID NO 10 < 211 > LENGTH : 1869 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 10 agcttttgga ccctcgtaca gaagctaata cgactcacta tagggaaata agagagaaaa 60 gaagagtaag aagaaatata agagccacca tggaagatgc gaagaacatc aagaagggac 120 ctgccccgtt ttaccctttg gaggacggta cagcaggaga acagctccac aaggcgatga 180 aacgctacgc cctggtcccc ggaacgattg cgtttaccga tgcacatatt gaggtagaca 240 tcacatacgc agaatacttc gaaatgtcgg tgaggctggc ggaagcgatg aagagatatg 300 gtcttaacac taatcaccgc atcgtggtgt gttcggagaa ctcattgcag tttttcatgo 360 cggtccttgg agcacttttc atcggggtcg cagtcgcgcc agcgaacgac atctacaatg 420 agcgggaact cttgaatagc atgggaatct cccagccgac ggtcgtgttt gtctccaaaa 480 aggggctgca gaaaatcctc aacgtgcaga agaagctccc cattattcaa aagatcatca 540 ttatggatag caagacagat taccaagggt tccagtcgat gtataccttt gtgacatcgc 600 atttgccgcc agggtttaac gagtatgact tcgtccccga gtcatttgac agagataaaa 660 ccatcgcgct gattatgaat tcctcgggta gcaccggttt gccaaagggg gtggcgttgc 720 cccaccgcac tgottgtgtg cggttctcgc acgctaggga tcctatcttt ggtaatcaga 780 tcattcccga cacagcaatc ctgtccgtgg taccttttca tcacggtttt ggcatgttca 840 cgactctcgg ctatttgatt tgcggtttca gggtcgtact tatgtatcgg ttcgaggaag 900 aactgttttt ttttctcatt agattgcctc tccaccttcc tcacgcccga 960 gtgccaacgc aatctgcatg gocaagcgct gcgatcctta ttctttgaag 1080 ccaaggtcgt agacctcgac acgggaaaaa ccctcggagt gaaccagagg ggcgagctct 1260 gcgtgagagg gccgatgatc atgtcaggtt acgtgaataa ccctgaagcg acgaatgcgc 1320 tgatcgacaa ggatgggtgg ttgcattcgg gagacattgc ctattgggat gaggatgagc 1380 acttctttat cgtagatega cttaagagct tgatcaaata caaaggctat caggtagcgc 1440 ctgccgagct cgagtcaatc ctgctccagc accccaacat tttcgacgcc ggagtggccg 1500 ggttgcccga tgacgacgcg ggtgagctgc cagcggccgt ggtagtcctc gaacatggga 1560 aaacaatgac cgaaaaggag atcgtggact acgtagcatc acaagtgacg actgegaaga 1620 aggtcccgaa ggcttgact gggaagcttg 1680 acgctcgcaa aatccgggaa atcctgatta aggcaaagaa aggcgggaaa atcgctgtct 1740 gataagctgc cttctgcggg gottgccttc tggccatgcc cttcttctct cccttgcacc 1800 ?? agggg gagatccttg caagattaca ctttgcgaaa tcgacactta agggggagcgccgcttagca cggaattcgg cagggatacg gggtgacgat aagccgggag tagtc tt agatccagtc ggccctcctt ttgataagta tgacctttcc aggaagtcggggaggcagtg ggctcacgga gacaacatcc ccgtcggaaa agtggtcccc 1020 1140 1200 US 10,898,574 B2 102 101 - continued tgtacctctt ggtctttgaa taaagcctga gtaggaaggc ggccgctcga gcatgcatct 1860 agagggccc 1869 < 210 > SEQ ID NO 11 < 211 > LENGTH : 930 < 212 > TYPE : DNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 11 tcaagctttt ggaccctcgt acagaagcta atacgactca ctatagggaa ataagagaga aaagaagagt aagaagaaat ataagagcca ccatggtgag caagggcgag gagctgttca 120 ccggggtggt gcccatcctg gtcgagctgg acggcgacgt aaacggccac aagttcagcg 180 tgtccggcga gggcgagggc gatgccacct acggcaagct gaccctgaag ttcatctgca 240 ccaccggcaa gctgcccgtg ccctggccca ccctcgtgac caccctgacc tacggcgtgc 300 agtgcttcag ccgctacccc gaccacatga agcagcacga cttcttcaag tocgccatgo 360 ccgaaggcta cgtccaggag cgcaccatct tcttcaagga cgacggcaac tacaagaccc 420 gcgccgaggt gaagttcgag ggcgacaccc tggtgaaccg catcgagctg aagggcatcg 480 acttcaagga ggacggcaac atcctggggc acaagctgga gtacaactac aacagccaca 540 acgtctatat catggccgac aagcagaaga acggcatcaa ggtgaacttc aagatccgcc 600 acaacatcga ggacggcagc gtgcagctcgccgaccacta ccagcagaac acccccatcg 660 gcgacggccc cgtgctgctg cccgacaacc actacctgag cacccagtcc gccctgagca 720 aagaccccaa cgagaagcgc gatcacatgg tcctgctgga gttcgtgacc gccgccggga 780 tcactctcgg catggacgag ctgtacaagt aagctgcctt ctgcggggct tgccttctgg 840 ccatgccctt cttctctccc ttgcacctgt acctcttggt ctttgaataa agcctgagta 900 ggaaggcggc cgctcgagca tgcatctaga 930 60 < 210 > SEQ ID NO 12 < 211 > LENGTH : 716 < 212 > TYPE : RNA < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 12 gggaaauaag agagaaaaga agaguaagaa gaaauauaag agccaccaug aacuuucucu 60 ugucaugggu gcacuggagc cuugcgcugc ugcuguaucu ucaucacgcu aaguggagcc 120 aggccgcacc cauggcggag gguggcggac agaaucacca cgaaguaguc aaauucaugg 180 acguguacca gaggucguau ugccauc?ga uugaaacucu uguggauauc uuucaagaau 240 accccgauga aaucgaguac auuuucaaac cgucgugugu cccucucaug aggugcgggg 300 gaugcugcaa ugaugaaggg uuggagugug uccccacgga ggagucgaau aucacaaugc 360 aaaucaugcg caucaaacca caucaggguc agcauauugg agagaugucc uuucuccago 420 acaacaaaug ugaguguaga ccgaagaagg accgagcccg acaggaaaac ccaugcggac 480 cgugcuccga gcggcgcaaa cacuuguucg uacaagaccc ccagacaugc aagugcucau 540 guaagaauac cgauucgcgg uguaaggcga gacagcugga auugaacgag cgcacgugua 600 ggugcgacaa gccuagacgg ugagcugccu ucugcggggc uugccuucug gccaugcccu 660 ucuucucucc cuugcaccug uaccucuugg ucuuugaaua aagccugagu aggaag 716 < 210 > SEQ ID NO 13 US 10,898,574 B2 104 103 - continued < 211 > LENGTH : 30 < 212 > TYPE : PRT < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 13 Met Ala Gly Pro Ala Thr Gln Ser Pro Met Lys Leu Met Ala Leu Gln 1 10 5 15 Leu Leu Leu Trp His Ser Ala Leu Trp Thr Val Gin Glu Ala 20 25 30 < 210 > SEQ ID NO 14 < 211 > LENGTH : 25 < 212 > TYPE : PRT < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 14 Met Met Pro Ser Ser Val Ser Trp Gly Ile Leu Leu Leu Ala Gly Leu 5 1 15 10 Cys Cys Leu Val Pro Val Ser Leu Ala 20 25 < 210 > SEQ ID NO 15 < 211 > LENGTH : 46 < 212 > TYPE : PRT < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 15 Met Gln Arg Val Asn Met Ile Met Ala Glu Ser Pro Ser Leu Ile Thr 5 1 10 15 Ile Cys Leu Leu Gly Tyr Leu Leu Ser Ala Glu Cys Thr Val Phe Leu 20 25 30 Asp His Glu Asn Ala Asn Lys Ile Leu Asn Arg Pro Lys Arg 35 40 45 < 210 > SEQ ID NO 16 < 211 > LENGTH : 21 < 212 > TYPE : PRT < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 16 Met Lys Gly Ser Leu Leu Leu Leu Leu Val Ser Asn Leu Leu Leu Cys 1 5 10 15 Gln Ser Val Ala Pro 20 < 210 > SEQ ID NO 17 < 211 > LENGTH : 24 < 212 > TYPE : PRT < 213 > ORGANISM : Homo sapiens < 400 > SEQUENCE : 17 Met Lys Trp Val Thr Phe Ile Ser Leu Leu Phe Leu Phe Ser Ser Ala 5 1 Tyr Ser Arg Gly Val Phe Arg Arg 20 10 15 US 10,898,574 B2 105 106 1. A method of producing a polypeptide of interest in a cell in a subject in need thereof, comprising administering to the modified uridine is modified on the major groove face of the uridine. 7. The pharmaceutical composition of claim 2 , wherein We claim : the subject a pharmaceutical composition comprising a 6. The pharmaceutical composition of claim 2 , wherein modified messenger ) suchmmRNA that thecomprises mmRNA 5 the modified uridine is a pyridine-4 -one ribonucleoside, is introduced into theRNA cell ,(mmRNA wherein the 5 - aza -uridine , 2 -thio - 5 - aza - uridine, 2 - thio -uridine, 4 -thio a translatable region encoding the polypeptide of interest and comprises the modified nucleoside 1 -methyl-pseudou ridine , and wherein the pharmaceutical composition com prises an effective amount of the mmRNA providing for 10 increased polypeptide production and substantially reduced innate immune response in the cell , as compared to a composition comprising a corresponding unmodified mRNA . 2. A pharmaceutical composition comprising: a plurality of lipid nanoparticles comprising a cationic 15 lipid , a sterol, and a PEG - lipid , wherein the lipid nanoparticles comprise an mRNA encoding a polypeptide, wherein the mRNA comprises one or more uridines, one or more cytidines , one or more adenosines, and one or more guanosines and 20 wherein substantially all uridines are modified uridines . 3. The pharmaceutical composition of claim 2 , wherein the plurality of lipid nanoparticles further comprise a phos phatidyl choline . pseudouridine, 2 -thio -pseudouridine, 5 -hydroxy - uridine, 3 -methyl -uridine, 5 -carboxymethyl -uridine, 1 -carboxym ethyl-pseudouridine, 5 -propynyl-uridine, 1 -propynyl pseudouridine, 5 - taurinomethyl -uridine, 1-taurinomethyl pseudouridine , 5 - taurinomethyl - 2 - thio - uridine, 1-taurino -4 thio -pseudouridine, 1 -methyl-pseudouridine, 4 - thio - 1 methyl -pseudouridine, 2 - thio - 1 -methyl- pseudouridine, 5. The pharmaceutical composition of claim 2 , wherein the plurality of lipid nanoparticles has a mean lipid to the mRNA further comprises an operably - linked signal 1 -methyl -1deaza -pseudouridine, 2- thio -1 -methyl-1 -deaza pseudouridine, dihydro - uridine, dihydro - pseudouridine, 2 - thio -dihydro -uridine, 2 - thio - dihydro -pseudouridine, 2 -methoxy -uridine, 2 -methoxy - 4 - thio -uridine, 4 -methoxy pseudouridine, 4 -methoxy - 2 -thio -pseudouridine , or pseudouridine. 8. The pharmaceutical composition of claim 2 , wherein the modified uridine is pseudouridine or 1 -methyl-pseudou ridine. 9. The pharmaceutical composition of claim 2 , wherein 4. The pharmaceutical composition of claim 2, wherein 25 the10.modified uridine is 1 -methyl -pseudouridine The pharmaceutical composition of claim. 2 , wherein the sterol is cholesterol. polynucleotide ratio ( wt /wt) of between 10 to 1 and 20 to 1 . sequence .

Disclaimer: Justia Dockets & Filings provides public litigation records from the federal appellate and district courts. These filings and docket sheets should not be considered findings of fact or liability, nor do they necessarily reflect the view of Justia.


Why Is My Information Online?