US00891. 1786B2

(12) United States Patent (10) Patent No.: US 8,911,786 B2 Desai et al. (45) Date of Patent: Dec. 16, 2014

(54) NANOPARTICLE COMPRISING RAPAMYCIN A61K 45/06 (2013.01); A61K 47/42 (2013.01); AND ALBUMINAS ANTICANCERAGENT A61N 5/10 (2013.01); A61N 7700 (2013.01) (75) Inventors: Neil P. Desai, Los Angeles, CA (US); USPC ...... 424/491; 424/489: 424/490; 424/500 Patrick Soon-Shiong, Los Angeles, CA (58) Field of Classification Search (US); Vuong Trieu, Calabasas, CA (US) USPC ...... 424/465-489, 490, 491, 500: 514/19.3 See application file for complete search history. (73) Assignee: Abraxis Bioscience, LLC, Los Angeles, CA (US) (56) References Cited (*) Notice: Subject to any disclaimer, the term of this patent is extended or adjusted under 35 U.S. PATENT DOCUMENTS U.S.C. 154(b) by 344 days. 5,206,018 A * 4/1993 Sehgal et al...... 424,122 5,362.478 A 11/1994 Desai et al. (21) Appl. No.: 12/530,188 5.439,686 A 8, 1995 Desai et al. 5,498.421 A 3, 1996 Grinstaffet al. (22) PCT Filed: Mar. 7, 2008 5,505,932 A 4/1996 Grinstaffet al. 5,508,021 A 4/1996 Grinstaffet al. (86). PCT No.: PCT/US2O08/OO3O96 5,512,268 A 4/1996 Grinstaffet al. 5,540,931 A 7/1996 Hewitt et al. S371 (c)(1), 5,560,933 A 10/1996 Soon-Shiong et al. (2), (4) Date: Mar. 4, 2010 5,635,207 A 6/1997 Grinstaffet al. 5,639,473 A 6/1997 Grinstaffet al. (87) PCT Pub. No.: WO2008/109163 5,650,156 A 7/1997 Grinstaffet al. PCT Pub. Date: Sep. 12, 2008 5,665,382 A 9, 1997 Grinstaffet al. 5,665,383 A 9, 1997 Grinstaffet al. (65) Prior Publication Data 5,916,596 A 6/1999 Desai et al. US 2010/O183728A1 Jul. 22, 2010 (Continued) FOREIGN PATENT DOCUMENTS Related U.S. Application Data (60) Provisional application No. 60/905,669, filed on Mar. EP 1683 S17 A1 T 2006 7, 2007, provisional application No. 60/905,734, filed JP 2004-525950 A 8, 2004 on Mar. 7, 2007, provisional application No. (Continued) 60/905,662, filed on Mar. 7, 2007, provisional application No. 60/905,735, filed on Mar. 7, 2007, OTHER PUBLICATIONS provisional application No. 60/905,672, filed on Mar. 7, 2007, provisional application No. 60/905,787, filed Adnot, S. etal. (Jan. 1991). “Loss of Endothelium-Dependent Relax on Mar. 7, 2007, provisional application No. ant Activity in the Pulmonary Circulation of Rats Exposed to Chronic 60/905,663, filed on Mar. 7, 2007, provisional Hypoxia.” J. Clin. Invest. 87(1): 155-162. application No. 60/905,767, filed on Mar. 7, 2007, provisional application No. 60/905,750, filed on Mar. (Continued) 7, 2007, provisional application No. 60/923,248, filed on Apr. 13, 2007, provisional application No. Primary Examiner — Michael G. Hartley 60/923,456, filed on Apr. 13, 2007. Assistant Examiner — Micah-Paul Young (51) Int. Cl. (74) Attorney, Agent, or Firm — Morrison & Foerster LLP A 6LX 9/50 (2006.01) A6 IK3I/675 (2006.01) A6 IK9/00 (2006.01) (57) ABSTRACT A 6LX 9/19 (2006.01) A 6LX 9/5 (2006.01) The present invention features methods for treating, stabiliz A6 IK3I/436 (2006.01) ing, preventing, and/or delaying cancer by administering A 6LX3/57 (2006.01) nanoparticles that comprise rapamycin or a derivative A6 IK3I/685 (2006.01) thereof. The invention also provides compositions (e.g., unit A6 IK 45/06 (2006.01) dosage forms) comprising nanoparticles that comprise a car A6 IK 47/42 (2006.01) rier protein and rapamycin or a derivative thereof. The inven A6IN 5/10 (2006.01) tion further provides combination therapy methods of treat A61N 7700 (2006.01) ing cancer comprising administering to an individual an (52) U.S. Cl. effective amount of nanoparticles that comprise rapamycin or CPC ...... A6 IK3I/675 (2013.01); A61 K9/0019 a derivative thereof and a second therapy. (2013.01); A61 K9/19 (2013.01); A61 K9/5169 (2013.01); A61 K3I/436 (2013.01); A61 K 31/517 (2013.01); A61 K3I/685 (2013.01); 37 Claims, 7 Drawing Sheets US 8,911,786 B2 Page 2

(56) References Cited 2013/0202709 A1 8, 2013 Desai et al. 2013/0209518 A1 8, 2013 Desai et al. U.S. PATENT DOCUMENTS 2013,0244952 A1 9/2013 Desai et al. 2013/0266659 A1 10, 2013 Desai et al. 5.997,904 A 12, 1999 Magdassi et al. 2013/0280336 A1 10, 2013 Desai et al. 6,096,331 A 8, 2000 Desai et al. 2013/0280337 A1 10, 2013 Desai et al. 6,506.405 B1 1, 2003 Desai et al. 2014, OO17315 A1 1/2014 Desai et al. 6,528,067 B1 3, 2003 Magdassi et al. 2014, OO1731.6 A1 1/2014 Desai et al. 6,537,579 B1 3, 2003 Desai et al. 2014, OO17323 A1 1/2014 Desai et al. 6,652,884 B2 11, 2003 Falciani 2014/OO23717 A1 1/2014 Desai et al. 6,749,868 B1 6, 2004 Desai et al. 2014/OO39069 A1 2/2014 Desai et al. 6,753,006 B1 6, 2004 Desai et al. 2014/OO39070 A1 2/2014 Desai et al. 7.771751 B2 8, 2010 Desai et al. 2014.0056986 A1 2/2014 Desai et al. 7,820,788 B2 10, 2010 Desai et al. 2014f0072630 A1 3/2014 Tao et al. 7,923,536 B2 4, 2011 Desai et al. 2014f0072631 A1 3/2014 Trieu et al. 7,981445 B2 T/2011 De et al. 2014f0072643 A1 3/2014 Desai et al. 8,034,375 B2 10, 2011 Desai et al. 2014/OO79787 A1 3/2014 Yeo et al. 8,034,765 B2 10, 2011 De et al. 2014/OO79788 A1 3/2014 Desai et al. 8,137,684 B2 3/2012 Desai et al. 2014/OO79793 A1 3/2014 Desai et al. 8,138,229 B2 3/2012 Desai et al. 2014f0080901 A1 3/2014 Desai et al. 8,257,733 B2 9, 2012 Desai et al. 2014/01342.57 A1 5, 2014 Desai et al. 8,268,348 B2 9, 2012 Desai et al. 8,314,156 B2 11, 2012 Desai et al. FOREIGN PATENT DOCUMENTS 2003, OO54042 A1 3, 2003 Liversidge et al. 2003/O1994.25 A1 10, 2003 Desai et al. WO WO-94, 18954 A1 9, 1994 2004/O126400 A1 T/2004 Iversen et al...... 424/400 WO WO-98,14174 A1 4f1998 2005/OOO40O2 A1 1/2005 Desai et al...... 514/2 WO WO-98, 14175 A1 4f1998 2005/OO38498 A1 2, 2005 Dubrow et al. WO WO-99/00113 A1 1, 1999 2005/01 19330 A1 6, 2005 Kao et al. WO WO-00,64437 A1 11, 2000 2005/O152979 A1 7/2005 Besman et al. WO WO-OOf71079 A2 11/2000 2005/02O3O13 A1 9, 2005 Soker et al. WO WO-OOf71079 A3 11, 2000 2005/0209266 A1 9, 2005 Garvey WO WO-01f89522 A1 11, 2001 2005/0244339 A1 11/2005 Jauernigh et al. WO WO-O2/O80975 A1 10, 2002 2006, OO73182 A1 4, 2006 Wong et al...... 424/426 WO WO-O2/O87545 A1 11, 2002 2006/0210638 A1 9, 2006 Liversidge et al. WO WO-03/096944 A1 11, 2003 2007/0066522 A1 3, 2007 McCormicket al...... 514/12 WO WO-2004/052401 A2 6, 2004 2007/0O87022 A1 4, 2007 Desai et al. WO WO-2004/0524O1 A3 6, 2004 2007/0092.563 A1 4, 2007 Desai et al. WO WO 2005.117952 A2 * 12/2005 2007/0093547 A1 4, 2007 Desai et al. WO WO-2006/053754 A1 5, 2006 2007/0116774 A1 5/2007 Desai et al. WO WO-2006/0892O7 A2 8, 2006 2007/0117744 A1 5/2007 Desai et al...... 514/11 WO WO-2006/0892O7 A3 8, 2006 2007/0129448 A1 6, 2007 Desai et al. WO WO-2006/08929.0 A1 8, 2006 2007/O166388 A1 7/2007 Desai et al. WO WO 20060892.90 A1 * 8, 2006 2008, OO63724 A1 3, 2008 Desai et al. WO WO-2006,124739 A1 11, 2006 2008. O153738 A1 6, 2008 Desai et al. WO WO-2007/027819 A2 3, 2007 2008. O161382 A1 T/2008 Desai et al. WO WO-2007/027941 A2 3, 2007 2008/0213370 A1 9, 2008 Desai et al. WO WO-2007/027941 A3 3, 2007 2008/0280939 A1 11, 2008 Izumo et al. WO WO-2008/O2.7055 A1 3, 2008 2008/028O987 A1 11, 2008 Desai et al. WO WO-2008/057562 A1 5, 2008 2009/0098.210 A1 4, 2009 Desai et al. WO WO-2008.076373 A1 6, 2008 2009/0175951 A1 T/2009 Liversidge WO WO-2008,109 163 A1 9, 2008 2009,0263.483 A1 10, 2009 Desai et al. WO WO-2008,137148 A2 11/2008 2009/0304805 A1 12, 2009 Desai et al. WO WO-2008,137148 A3 11 2008 2010.0035.800 A1 2, 2010 Desai et al. WO WO-2008,150532 A1 12/2008 2010.0048499 A1 2, 2010 Desai et al. 2010/01 12077 A1 5, 2010 Desai et al. OTHER PUBLICATIONS 2010, 0166869 A1 T/2010 Desai et al. 2010/0215751 A1 8, 2010 Desai et al. Al Housseini. A et al. (Mar. 1, 2008). “A Phase II. Non-Randomized 2010, O297243 A1 11, 2010 Desai et al. Study of Abraxane plus in Patents With Recurrent Plati 2011/0052708 A1 3, 2011 Soon-Shiong et al. 2011 OO64812 A1 3, 2011 Bahl et al. num-Sensitive Ovarian or Primary Peritoneal Cancer: Evaluation of 2011 0118342 A1 5, 2011 De et al. the Response and Survival and Progression-Free Survival.” Gyneco 2011 O151012 A1 6, 2011 Desai et al. logic Oncology, 108(Suppl. 1): S129-S130, Abstract No. 293. 2011/O165256 A1 T/2011 Desai et al. Carmeliet, P. et al. (Sep. 14, 2000). "Angiogenesis in Cancer and 2011 0196026 A1 8, 2011 De et al. Other Diseases.” Nature 407(6801):249-257. 2011/030.1248 A1 12, 2011 Desai et al. Carter, D. C. et al. (1994). "Structure of Serum Albumin.” Adv. 2012fOOO4177 A1 1, 2012 Desai et al. Protein. Chen. 45:153-203. 2012,00705O2 A1 3/2012 Desai et al. 2012/0076862 A1 3/2012 Desai et al. Chauhan, D. etal. (Feb. 1, 1996). “ Cell Adhesion 2012/O128732 A1 5, 2012 Trieu et al. Induced Interleukin-6 Express in Bone Marrow Stromal Cells 2012/O1897O1 A1 T/2012 Desai et al. Involves Activation of NF-Kappa B.” Blood 87(3): 1104-1112. 2012fO23.1082 A1 9, 2012 Desai et al. Curry, S. et al. (Sep. 1998). “Crystal Structure of Human Serum 2012,0283205 A1 11, 2012 Desai et al. Albumin Complexed with Fatty Acid Reveals an Asymmetric Distri 2013,0045240 A1 2, 2013 Tao et al. bution of Binding Sites.” Nat. Struct. Biol. 5(9):827-835. 2013,007 1438 A1 3/2013 Desai et al. Curry, S. et al. (1999). "Fatty Acid Binding to Human Serum Albu 2013/01 15296 A1 5, 2013 Yeo et al. min: New Insights from Crystallographic Studies.” Biochim. 2013,0195922 A1 8, 2013 Desai et al. Biophys. Acta 1441:131-140. 2013,0195983 A1 8, 2013 Desai et al. De Tapas, K. et al. (Apr. 17, 2007) “Nanoparticle Albumin-Bound 2013,0195984 A1 8, 2013 Desai et al. (nab) Rapamycin as an Anticancer Agent.” Proceedings of the Annual US 8,911,786 B2 Page 3

(56) References Cited Therasse, P. et al. (Feb. 2, 2000). “New Guidelines to Evaluate the Response to Treatment in Solid Tumors.” J. Natl. Cancer Inst. OTHER PUBLICATIONS 92(3):205-216. Trieu, V. etal. (Dec. 2007). “mTORInhibitor Nanoparticle Albumin Meeting of the American Association for Cancer Research, held on Bound (nab) Rapamycin is Effective in a Breast Cancer Xenograft Apr. 14, 18, 2007, in Los Angeles, California 48(1): 1117. Abstract Model.” Breast Cancer Research and Treatment, 30th Annual San No. 4719. Antonio Breast Cancer Symposium; San Antonio, Texas, USA, held Ellerby, H. M. et al. (Sep. 1999). "Anti-Cancer Activity of Targeted on Dec. 13-16, 2007 106(supplement 1):S268, Abstract 6063. Pro-Apoptotic Peptides.” Nat. Med. 5(9): 1032-1038. Uchiyama, H. etal. (Dec. 15, 1993). “Adhesion of Human Myeloma Fehske, K. J. et al. (Apr. 1, 1981). “The Location of Drug Binding Derived Cell Lines to Bone Marrow Stromal Cells Stimulates Sites in Human Serum Albumin.” Biochem. Pharmacol. 30(7):687 Interleukin-6 Secretion.” Blood 82(12):3712-3720. 692. Vorum, H. (Nov. 1999). “Reversible Ligand Binding to Human Gartner, S. et al. (Aug. 1980). “Long-Term Culture of Human Bone Serum Albumin. Theoretical and Clinical Aspects.” Dan. Med. Bull. Marrow Cells.” Proc. Natl. Acad. Sci. USA 77(8):4756-4759. 46(5):379-399. Gupta, D. etal. (Dec. 2001). “Adherence of Multiple Myeloma Cells Zaiman, A. etal. (Nov. 2005). "One HundredYears of Research in the to Bone Marrow Stromal Cells Upregulates Vascular Endothelial Pathogenesis of Pulmonary Hypertension.” Am. J. Respir: Cell Mol. Growth Factor Secretion: Therapeutic Applications.” Leukemia Biol. 33(5):425-431. 15(12): 1950-1961. Zhou, H. al. (Jan. 2006). “Heme Oxygenase-1 Mediates the Protec Hawkins, M. J. et al. (May 22, 2008, e-pub. Feb. 7, 2008). “Protein tive Effects of Rapamycin in Monocrotaline-Induced Pulmonary Nanoparticles as Drug Carriers in Clinical Medicine.” Advanced Hypertension.” Lab. Investigation 86(1):62-71. Drug Delivery Reviews 60(8):876-885. International Search Report mailed on Jul. 22, 2008, for PCT Patent He, X. M. et al. (Jul. 16, 1992). "Atomic Structure and Chemistry of Application No. PCT/US2008/003096 filed on Mar. 7, 2008, pub Human Serum Albumin.” Nature 358(6383): 209-215. lished as WO 2008/109 163 on Sep. 12, 2008, 3 pages. Herzog, T. J. (Nov. 15, 2004). “Recurrent Ovarian Cancer. How Written Opinion mailed on Jul. 22, 2008, for PCT Patent Application Important is it to Treat to Disease Progression?” Clin. Cancer Res. No. PCT/US2008/003096 filed on Mar. 7, 2008, published as WO 10:7439-7449. 2008/109 163 on Sep. 12, 2008, 10 pages. Hideshima, T. et al. (May 15, 2006). “Perifosine, An Oral Bioactive International Search Report mailed on Dec. 22, 2008, for PCT Patent Novel Alkylphospholipid, Inhibits Akt and Induces in Vitro and in Application No. PCT/US2008/005792 filed on May 5, 2008, pub Vivo Cytotoxicity in Human Multiple Myeloma Cells.” Blood lished as WO 2008/137148 on Nov. 13, 2008, 7 pages. 107(10):4053-4062. Written Opinion mailed on Dec. 22, 2008, for PCT Patent Applica Icon et al. (Jun. 21, 2003). “ Plus Platinum-Based Chemo tion No. PCT/US2008/005792 filed on May 5, 2008, published as therapy Versus Conventional Platinum-Based in WO 2008/150531 on Nov. 13, 2008, 10 pages. Women with Relapsed Ovarian Cancer: The ICON4/AGO-OVAR International Search Report mailed on Oct. 9, 2008, for PCT Patent 2.2 Trial.” Lancet 361 (93.75): 2099-2106. Application No. PCT/US2008/007024 filed on Jun. 2, 2008, pub Koshkina, N. V. et al. (2003). “ and Lung Cancer. lished as WO 2008/150532 on Dec. 11, 2008, 3 pages. Improved Delivery Systems by Aerosol.” Current Cancer Drug Tar Written Opinion mailed on Oct. 9, 2008, for PCT Patent Application gets 3(4):251-264. No. PCT/US2008/007024 filed on Jun. 2, 2008, published as WO Kragh-Hansen, U. (Feb. 1990). "Structure and Ligand Binding Prop 2008/150532 on Dec. 11, 2008, 6 pages. erties of Human Serum Albumin.” Dan. Med. Bull, 37(1)57-84. European Office Action mailed on Jun. 30, 2011, for European Patent Micha, J. P. et al. (Feb. 2006, e-pub. Oct. 15, 2005). “Abraxane in the Application No. 08 768 111.0 filed on Jun. 2, 2008, 5 pages. Treatment of Ovarian Cancer: The Absence of Hypersensitivity Non Final Office Action mailed on Sep. 27, 2011, for U.S. Appl. No. Reactions.” Gynecologic Oncology 100(2):437-438. 12/832,876, filed Jul. 8, 2010, for Desai et al., 16 pages. Mitsiades, C. S. et al. (Aug. 1, 2001). “Trail/Apo2L Ligand Selec Non Final Office Action mailed on Nov. 21, 2011, for U.S. Appl. No. tively Induces Apoptosis and Overcomes Drug Resistance in Mul 12,422, 192, filed Apr. 10, 2009, 20 pages. tiple Myeloma: Therapeutic Applications.” Blood 98(3):795-804. U.S. Appl. No. 09/937,840, filed Jan. 28, 2002, for Desai et al. Mitsiades, N. et al. (Oct. 29, 2002). “Molecular Sequelae of Protea U.S. Appl. No. 12/479,710, filed Jun. 5, 2009, for Desai et al. some Inhibition in Human Multiple Myeloma Cells.” Proc. Natl. U.S. Appl. No. 13/228,323, filed Sep. 8, 2011, for Desai et al. Acad. Sci. USA99(22): 14374-14379. U.S. Appl. No. 13/263,723, internationally filed Apr. 9, 2010, for Mitsiades, N. etal. (May 15, 2003). “Molecular Sequelae of Histone Desai et al. Deacetylase Inhibition in Human Malignant B Cells.” Blood U.S. Appl. No. 13/133,367, internationally filed Dec. 11, 2009, for 101 (10):4055-4062. Trieu et al. Mitsiades, N. et al. (Mar. 15, 2003). “The U.S. Appl. No. 13/255,893, internationally filed Mar. 12, 2010, for PS-341 Potentiates Sensitivity of Multiple Myeloma Cells to Con Desai et al. ventional Chemotherapeutic Agents: Therapeutic Applications.” U.S. Appl. No. 13/073,824, filed Mar. 28, 2011, for Desai et al. Blood 101(6):2377-2380. U.S. Appl. No. 13/073,861, filed Mar. 28, 2011, for Desai et al. Muggia, F. M. (Dec. 2006). “New and Emerging Intraperitoneal (IP) Ballou, L. M. et al. (Jun. 27, 2003). “Activated Go inhibits p110O. Drugs for Ovarian Cancer Treatment.” Seminars in Oncology Phosphatidylinositol 3-Kinase and Akt.” J. Biol. Chem. 33(6)(Suppl. 12):S18-S24. 278(26):23472-23479. Nishimura.T. etal. (Feb. 2001). “40-O-(2-Hydroxyethyl)-rapamycin De Mulder, P.H. et al. (2004). “Current Treatment of Renal Cell Attenuates Pulmonary Arterial Hypertension and Neointimal Forma Carcinoma.” Ann. Oncol. 15(Suppl. 4):319-328. tion in Rats.” Am. J. Respir: Crit. Care Med. 163(2):498-502. Desai, N. et al. (Dec. 2009). Combination Regimens of nab Paddenberg, R. et al. (Feb. 24, 2007). “Rapamycin Attenuates Rapamycin (ABI-009) Effective Against MDA-MB-231 Breast-Tu Hypoxia-Induced Pulmonary Vascular Remodeling and Right mor Xenografts, Cancer Res.69(24), Supplement 3, ABS 6106,32nd Ventricular Hypertrophy in Mice.” Respiratory Research 8(1):15, Annual San Antonio Breast Cancer Symposium, San Antonio, Texas, total of 12 pages. USA, located at shttp://cancerres.aacrjournals.org/cgi/content/ab Rustin, G. J. et al. (Jun. 1, 2004). “Use of CA-125 in stract/69/24 Meeting Abstracts/6106, one page. Evaluation of New Therapeutic Drugs for Ovarian Cancer.” Clin. Hung, G.Y. et al. (Jan. 2002). “Nonmyeloablative Allogeneic Bone Cancer Res. 10(11):3919-3926. Marrow Transplantation for Orbital Granulocytic Sarcoma Associ Sugio, S. et al. (Jun. 1999). "Crystal Structure of Human Serum ated with tC8:21)(cq22:cq22) in Acute Myeloid Leukemia.” Bone Mar Albumin at 2.5 A Resolution.” Protein. Eng. 12(6):439-446. row Transplant. 29(1):67-70. Tai, Y-T. et al. (Feb. 21, 2000). “Isolation and Characterization of Kim. D. et al. (Dec. 1, 2005). “Targeting the Phosphatidylinositol-3 Human Multiple Myeloma Cell Enriched Populations.” J. Immunol. Kinase / Akt Pathway for the Treatment of Cancer.” Curr: Opin. Methods 235(1-2): 11-19. Invest. Drugs. 6(12): 1250-1258. US 8,911,786 B2 Page 4

(56) References Cited Kawagoe, H. et al. (Jan. 2003). “Weekly Paclitaxel Infusion in Patients with Recurrent Ovarian Cancer—A Pilot Study.” Japanese OTHER PUBLICATIONS Journal of Cancer and Chemotherapy 30(1): 151-154. (English Abstract). Konski, A. et al. (Jan. 1, 2006, e-ub. Dec. 1, 2005). “Long-Term Pfizer. (2012). Patient Information Leaflet included with Hormone Therapy and Radiation is Cost-Effective for Patients With Rapamune(R) (Sirolimus) Oral Solution and Tablets Packages Insert Locally Advanced Prostate Carcinoma.” Cancer 106(1):51-57. LAB-0579-1.0, instructions approved by the U.S. Food and Drug Liu, X. Y. et al. (Oct. 2006). “A Mini-Review of Targeting Gene Administration, published under “Full Prescribing Information.” at Virotherapy of Cancer.” Chinese Journal of Cancer 25(10): 1320 http://labeling.pfizer.com/showlabeling.aspx?id=139, instructions 1322. revised Dec. 2012 by Pfizer distributed by Wyeth Pharmaceuticals, Liu, L. et al. (Nov. 9, 2006). “Rapamycin Inhibits Cell Motility by Inc., 53 pages. Suppression of mTOR-Mediated S6K1 and 4E-BP1 Pathways.” Rose, P.G. et al. (1998). "Second-Line Therapy With Paclitaxel and Oncogene 25(10):7029-7040. Carboplatin for Recurrent Disease Following First-Line Therapy Luan, F. L. et al. (May 27, 2022). “Rapamycin Blocks Tumor Pro With Paclitaxel and Platinum in Ovarian or Peritoneal Carcinoma.” gression: Unlinking Immunosuppression From Antitumor Efficacy.” Journal of Clinical Oncology 16(4): 1494-1497. raba, S. et al. (Mar. 2004). “Recurrent Ovarian Cancer Peritonitis Transplantation 73(10) 1565-1572. reated with Weekly Paclitaxel Infusion: A Clinicopharmacological Muramatsu, T. etal. (Jul. 1, 2006). “Treatment Strategy for Recurrent tudy.” Japanese Journal of Cancer and Chemotherapy 31(3):449 and Refractory Epithelial Ovarian Cancer: Efficacy of High-Dose s 51. (English Abstract). Chemotherapy With Hematopoietic StemCell Transplantation.” Acta . Appl. No. 13/649.987, filed Oct. 11, 2012, for Desai et al. Histochem. Cytochem. 39(3):61-67. . Appl. No. 13/777,980, filed Feb. 26, 2013, for Desai et al. Sarbassov, D.D. etal. (Apr. 21, 2006). “Prolonged Rapamycin Treat . Appl. No. 13/777,988, filed Feb. 26, 2013, for Desai et al. ment Inhibits mTORC2 Assembly and Akt/PKB.” Mol. Cell. . Appl. No. 13/564,633, filed Aug. 1, 2012, for Desai et al. 22(2): 159-168. . Appl. No. 13/585,696, filed Aug. 14, 2012, for Desai et al. Sato, A. et al. (2006). “Prophylaxis of Mucosal Toxicity by Oral . Appl. No. 13/743,212, filed Jan. 1, 2013, for Desai et al. Propantheline and Cryotherapy in Children With Malignancies . Appl. No. 13/776,481, filed Feb. 25, 2013, for Desai et al. Undergoing Myeloablative Chemo-Radiotherapy,” Tohoku J. Exp. . Appl. No. 13/776,484, filed Feb. 25, 2013, for Desai et al. Med. 210(4):315-320. . Appl. No. 13/779,625, filed Feb. 27, 2013, for Desai et al. Wu, F. et al. (Jul. 2005). “Extracorporeal High Intensity Focused . Appl. No. 13/779,624, filed Feb. 27, 2013, for Desai et al. Ultrasound Treatment for Patients With Breast Cancer. Breast Can . Appl. No. 13/779,621, filed Feb. 27, 2013, for Desai et al. cer Res. Treat. 92(1):51-60. . Appl. No. 13/781,482, filed Feb. 28, 2013, for Desai et al. Wullschleger, S. et al. (2006). “TOR Signalling in Growth and . Appl. No. 13/781,479, filed Feb. 28, 2013, for Desai et al. Metabolism.” Cell 124(3):471-484. . Appl. No. 13/781,489, filed Feb. 28, 2013, for Trieu et al. European Search Report mailed on Sep. 7, 2012, for European Patent . Appl. No. 13/781.487, filed Feb. 28, 2013, for Tao et al. Application No. 12152463. 1, filed on Mar. 7, 2008, published on . Appl. No. 13/781,480, filed Feb. 28, 2013, for Yeo et al. Aug. 1, 2012, as EP 2 481 409, 7 pages. . Appl. No. 13/782,990, filed Mar. 1, 2013, for Desai et al. European Search Report mailed on Sep. 11, 2012, for European . Appl. No. 13/782.988, filed Mar. 1, 2013, for Desai et al. Patent Application No. 08767585.6, filed on Mar. 5, 2008, published . Appl. No. 13/701,003, filed May 20, 2011, for Desai et al. on Jan. 27, 2010, as EP 2 146 707, 4 pages. . Appl. No. 13/782,984, filed Mar. 1, 2013, for Desai et al. European Office Action mailed on Jul. 5, 2012, for European Patent . Appl. No. 13/701,002, filed May 20, 2011, for Desai et al. Application No. 087681 11.0 filed on Jun. 2, 2008, 3 pages. . Appl. No. 13/783,122, filed Mar. 1, 2013, for Desai et al. European Office Action mailed on Sep. 4, 2012, for European Patent . Appl. No. 13/701,001, filed May 20, 2011, for Desai et al. Application No. 08726.605.2 filed on Mar. 7, 2008, 7 pages. . Appl. No. 13/782,992, filed Mar. 1, 2013, for Desai et al. European Search Report and European Search Opinion mailed on . Appl. No. 13/794,705, filed Mar. 12, 2013, for Desai et al. Sep. 17, 2012, for European Patent Application No. 12152455.7, filed . Appl. No. 13/791,841, filed Mar. 12, 2013, for Desai et al. on Mar. 7, 2008, published on Aug. 1, 2012, as EP2481 402.9 pages. . Appl. No. 13/794,480, filed Mar. 12, 2013, for Desai et al. Final Office Action mailed on Apr. 27, 2012, for U.S. Appl. No. . Appl. No. 13/794,486, filed Mar. 12, 2013, for Heise et al. 12/832,876, filed Jul. 8, 2010, for Desai et al., 14 pages. . Appl. No. 13/794,712, filed Mar. 11, 2013, for Pierce et al. Final Office Action mailed on May 22, 2012, for U.S. Appl. No. Aapro, M. etal. (Apr. 2014). “Adverse Event Management in Patients 12,422, 192, filed Apr. 10, 2009, 26 pages. With Advanced Cancer Receiving Oral Everolimus: Focus on Breast U.S. Appl. No. 09/446,783, filed May 16, 2000, for Desai et al. Cancer.” Annals of Oncology 25(4):763-773. U.S. Appl. No. 13/392,501, internationally filed Aug. 25, 2010, for AFINITOR(R) Insert. (Feb. 2014). “Highlights of Prescribing Infor Tao et al. mation.” AFINITORR) pp. 1-41. U.S. Appl. No. 13/368,250, filed Feb. 7, 2012, for Desai et al. Amato, R. J. et al. (Jun. 1, 2009, e-pub. Mar. 20, 2009). "A Phase 2 U.S. Appl. No. 13/368,297, filed Feb. 7, 2012, for Desai et al. Study With a Daily Regimen of the Oral mTOR Inhibitior RAD001 U.S. Appl. No. 13/408,994, filed Feb. 29, 2012, for De et al. (Everolimus) in Patients With Metastatic Clear Cell Renal Cell Can U.S. Appl. No. 13/423,095, filed Mar. 16, 2012, for Desai et al. cer, Cancer 115:2438-2446. Duan Qiong-wen et al. "Rapamycin Associated Molecular Target Atkins, M.B. et al. (Mar. 1, 2004). “Radomized Phase II Study of mTOR Development of Novel Antineoplastic Drugs.” translated as Multiple Dose Levels of CCI-779, a Novel Mammalian Target of “Molecular Target of Rapamycin mTOR Associated With the Devel Rapamycin Kinase Inhibitor, in Patients With Advanced Refractory opment of New Anticancer Drugs,” Journal of Medicine Research Renal Cell Carcimona.” Journal of Clinical Oncology 22(5):909 35(4):55-58, (Apr. 30, 2006). 918. Noh, W-C. et al. (Feb. 1, 2004). “Determinants of Rapamycin Sen Baselga, J. et al. (Feb. 9, 2012, e-pub. Dec. 7, 2011). “Everolimus in sitivity in Breast Cancer Cells.” Clinical Cancer Research 10:1013 Postmenopausal Hormone-Receptor-Positive Advanced Breast Can 1023. cer.” The New England Journal of Medicine 3.66(6):520-529. Beppo, M. et al. (May 2001). “Weekly Paclitaxel and in Baselga, J. et al. (Jun. 1, 2009, e-pub. Jan. 12, 2009). "Phase II Recurrent Ovarian Cancer.” Japanese Journal of Cancer and Che Randomized study of Neoadjuvant Everolimus Plus Letrozole Com motherapy 28(50):643-648. (English Abstract). pared With Placebo Plus Letrozole in Patients With Estrogen Recep Gonzalez-Martin, A.J. et al. (2005). “Randomized Phase II Trial of tor-Positive Breast Cancer.” Journal of Clinical Oncology Carboplatin Versus Paclitaxel and Carboplatin in Platinum-Sensitive 27(16):2630-2637. Recurrent Advanced Ovarian Carcinoma: A GEICO (Grupo Español Bryce, A.H. et al. (Oct. 2012). "Phase I Study of Temsirolimus in de Investigacion en Cancer de Ovario) Study.” Annuals of Oncology Combination With EKB-569 in Patients With Advanced Solid 16(5):749-755. Tumors.” Invest. New Drugs 30(5):1-16. US 8,911,786 B2 Page 5

(56) References Cited Hudes, G. et al. (May 31, 2007). "Temsirolimus, Interferon Alfa, or Both for Advanced Renal-Cell Carcinoma.” The New England Jour OTHER PUBLICATIONS nal of Medicine 356(22):2271-2281. Javle, M.M. et al. (2010). "Inhibition of the Mammalian Target of Buckner, J.C. et al. (2010, e-pub. May 5, 2009). “Phase I, Rapamycin (mTOR) in Advanced Pancreatic Cancer: Results of Two Pharmacokinetic Study of Termsirolimus Administered Orally to Phase II Studies.” BMC Cancer 10(368)1-7. Patients With Advanced Cancer.” Invest. New Drugs 28:334-342. Kita, T. et al. (2004). The Effect of Single Weekly Paclitaxel in Campone, M. et al. (2009, e-pub. Jan. 6, 2009). "Safety and Heavily Pretreated Patients with Recurrent or Persistent Advanced Pharmacokinetics of Paclitaxel and the Oral mTOR Inhibitor Ovarian Cancer, Gynecologic Oncology 92:813-818. Everolimus in Advanced Solid Tumuors.” British Journal of Cancer Margolin, K. et al. (Sep. 1, 2005, e-pub. Jul. 8, 2005). “CCI-779 in 100(2):315-321. Metastatic Melanoma. A Phase II Trial of the California Cancer Chan, S. et al. (Aug. 10, 2005). "Phase II Study of Temsirolimus Consortium.” Cancer 104(5):1045-1048. (CCI-779), A Novel Inhibitor of mTOR, in Heavily Pretreated Martins, F. et al. (2013, e-pub. Jan. 9, 2013). “A Review of Oral Patients With Locally Advanced or Metastatic Breast Cancer.” Jour Toxicity Associated With mTOR Inhibitor Therapy in Cancer nal of Clinical Oncology 23(23):5314-5322. Patients.” Oral Oncology 49:293–298. Chawla, S.P. et al. (Jan. 1, 2012, e-pub. Nov. 7, 2011). “Phase II Study Meier-Kriesche, H-U. et al. (2000). "Toxicity and Efficacy of of the Mammalian Target of Rapamycin Inhibitor Ridaforolimus in Sirolimus: Relationship to Whole-Blood Concentrations.” Clinical Patients With Advanced Bone and Soft Tissue Sarcomas,” Journal of Therapeutics 22(Suppl. B):B93-B100. Clinical Oncology 30(1):78-84. Milton, D.T. etal. (Aug. 1, 2007, e-pub. Jun. 18, 2007). "Phase 1 Trial Cohen, E.E.W. et al. (Sep. 1, 2012, e-pub. Aug. 7, 2012). "Phase I of Everolimus and Gefitinib in Patients With Advanced Nonsmall Studies of Sirolimus Alone or in Combination With Pharmacokinetic Cell Lung Cancer.” Cancer 110(3):599-605. Modulators in Advanced Cancer Patients.” Clin. Cancer Res. Mita, M.M. et al. (Jan. 20, 2008). “Phase I Trial of the Novel Mam 18(17):4785-4793. malian Target of Rapamycin Inhibitor Deforolimus (AP23573; Deenen, M.J. et al. (2012, e-pub. Aug. 2, 2011). “Phase I and MK-8669) Administered Intravenously Daily for 5 Days Every 2 Pharmacokinetic Study of and the Oral mTOR Inhibi Weeks to Patients With Advanced Malignancies,” Journal of Clinical tor Everolimus in Patients with Advanced Solid Malignancies.” Oncology 26(3):361-367. Invest. New Drugs 30:1557-1565. Motzer, R. J. et al. (Sep. 1, 2007). “Phase I/II Trial of Temsirolimus Doi, T. et al. (Apr. 10, 2010, e-pub. Mar. 15, 2010). “Multicenter Combined With Interferon Alfa for Advanced Renal Cell Carci Phase II Study of Everolimus in Patients With Previously Treated noma.” Journal of Clinical Oncology 25(25):3958-3965. Metastatic Gastric Cancer.” Journal of Clinical Oncology Motzer, R.J. et al. (Sep. 15, 2010, e-pub. Jun. 14, 2010). “Phase 3 28(11):1904-1910. Trial of Everolimus for Metastatic Renal Cell Carcinoma, Cancer Duran, I. et al. (2006, e-pub. Oct. 10, 2006). “A Phase II Clinical and 116:4256-4265. Pharmacodynamic Study of Temsirolimus in Advanced Naing, A. etal. (Sep. 15, 2011, e-pub. Jul. 12, 2011). “Phase I Trial of Neuroendocrine Carcinomas.” British Journal of Cancer Cixutumumab Combined With Temsirolimus in Patients With 95(9): 1148-1154. Advanced Cancer.” Clin. Cancer Res. 17(18):6052-6060. Ellard, S.L. et al. (Sep. 20, 2009, e-pub. Aug. 17, 2009). “Random O'Donnell, A. et al. (Apr. 1, 2008, e-pub. Mar. 10, 2008). “Phase I ized Phase II Study Comparing Two Schedules of Everolimus in Pharmacokinetic and Pharmacodynamic Study of the Oral Mamma Patients. With Recurrent/Metastatic Breast Cancer: NCIC Clinical lian Target of Rapamycin Inhibitior Everolimus in Patients With Trials Group IND. 163,” Journal of Clinical Oncology 27(27):4536 Advanced Solid Tumors,” Journal of Clinical Oncology 4541. 26(10): 1588-1595. Farag, S.S. et al. (Nov. 2009). "Phase II Trial of Temsirolimus in Okuno, S. etal. (Aug. 1, 2011, e-pub. Feb. 1, 2011). A Phase 2 Study Patients with Relapsed or Refractory Multiple Myeloma.” Leuk. Res. of Temsirolimus (CCI-779) in Patients With Soft Tissue Sarcomas, 33(11): 1475-1480. Concer 117:3468-3475. Fouladi, M. et al. (Oct. 20, 2007). “Phase I Study of Everolimus in Perotti, A. et al. (Oct. 20, 2010, e-pub. Sep. 20, 2010). “Phase IB Pediatric Patients With Refractory Solid Tumors,” Journal of Clinical Study of the mTOR Inhibitor Ridaforolimus With Capecitabine.” Oncology 25(30):4806-4812. Journal of Clinical Oncology 28(30):4554-4561. Fujisaka, Y. et al. (2010, e-pub. Apr. 29, 2010). “A Phase 1 Clinical Pham. P-T.T. et al. (Apr. 27, 2004). “Sirolimus-Associated Pulmo Study of Temsirolimus (CCI-779) in Japanese Patients With nary Toxicity.” Transplantation 77(8): 1215-1220. Advanced Solid Tumor.” Jpn. J. Clin. Oncol. 40(8): 732-738. Punt, C.J.A. et al. (2003). “Phase I and Pharmacokinetic Study of Galanis, E. et al. (Aug. 10, 2005). "Phase II Trial of Temsirolimus CCI-779, a Novel Cytostatic Cell-Cycle Inhibitor, in Combination (CCI-779) in Recurrent Glioblastoma Multiforme: A North Central With 5- and Leucovorin in Patients With Advanced Solid Cancer Treatment Group Study,” Journal of Clinical Oncology Tumors.” Annals of Oncology 14:931-937. 23(23):5294-5304. Quek, R. et al. (Feb. 15, 2011, e-pub. Dec. 22, 2010). “Combination Gonzalez-Angulo, A.M. et al. (Oct. 1, 2013). “Weekly nab mTOR and IGF-1 R Inhibition: Phase I Trial of Everolimus and Rapamycin in Patients With Advanced Nonhematologic Malignan Figitumumab in Patients With Advanced Sarcomas and Other Solid cies: Final Results of a Phase I Trial.” Clinical Cancer Research Tumors.” Clin. Cancer Res. 17(4):871-879. 19(19):5474-5484. Raymond, E. etal. (Jun. 15, 2004). “Safety and Pharmacokinetics of Hainsworth, J.D. et al. (May 1, 2010, e-pub. Apr. 5, 2010). "Phase II Escalated Doses ofWeekly Intravenous Infusion of CCI-779, a Novel Trial of Bevacizumab and Everolimus in Patients With Advanced mTORInhibitor, in Patients With Cancer.” Journal of Clinical Oncol Renal Cell Carcinoma.” Jounal of Clinical Oncology 28(13):2131 ogy 22(12):2336-2347. 2136. Rizzieri, D.A. et al. (May 1, 2008). “A Phase 2 Clinical Trial of Hartford, C.M. et al. (Feb. 15, 2009). “A Phase I Trial to Determine Deforolimus (AP23573, MK-8669), A Novel Mammalian Target of the Safety, Tolerability, and Maximum Tolerated Dose of Rapamycin Inhibitor, in Patients With Relapsed or Refractory Hema Deforolimus in Patients With Advanced Malignancies.” Clin. Cancer tologic Malignancies.” Clin. Cancer Res. 14(9):2756-2762. Res. 15(4): 1428-1434. Seront, E. et al. (Oct. 2012, e-pub. Apr. 3, 2012). "Phase II Study of Hess, G. et al. (Aug. 10, 2009, e-pub. Jul. 6, 2009). "Phase III Study Everolimus in Patients With Locally Advanced or Metastatic Transi to Evaluate Temsirolimus Compared With Investigator's Choice tional Cell Carcinoma of the Urothelial Tract: Clinical Activity, Therapy for the Treatment of Relapsed or Refractory Mantle Cell Molecular Response, and Biomarkers.” Annals of Oncology Lymphoma.” Journal of Clinical Oncology 27(23):3822-2829. 23(10):2663-2670. Hidalgo, M. et al. (Oct. 1, 2006). “A Phase I and Pharmacokinetic Sessa, C. et al. (Jun. 2010, e-pub. Nov. 9, 2009). “Phase 1b Study of Study of Temsirolimus (CCI-779) Administered Intravenously Daily Weekly Mammalian Target of Rapamycin Inhibitor Ridaforolimus for 5 Days Every 2 Weeks to Patients With Advanced Cancer.” Clin. (AP23573; MK-8669) With Weekly Paclitaxel.” Annals of Oncology Cancer Res. 12(19):5755-5763. 21(6): 1315-1322. US 8,911,786 B2 Page 6

(56) References Cited Torisel(R). (May 2014). “Highlights of Prescribing Information.” located at . OTHER PUBLICATIONS last visited on May 15, 2014, 21 pages. Witzig, T.E. et al. (Aug. 10, 2005). "Phase II Trial of Single-Agent Sivendran, S. et al. (2014). “Metabolic Complications With the Use Temsirolimus (CCI-779) for Relapsed Mantle Cell Lymphoma.” of mTORInhibitors for Cancer Therapy,” Cancer Treatment Reviews Journal of Clinical Onology 23(23):5347-5356. 40:190-196. Wolpin, B.M. etal. (Jan. 10, 2009, e-pub. Dec. 1, 2008). “Oral mTOR Slomovitz, B.M. etal. (Dec. 1, 2010, e-pub. Aug. 2, 2010). “A Phase Inhibitor Everolimus in Patients With -Refractory Meta 2 Study of the Oral Mammalian Target of Rapamycin Inhibitor, static Pancreatic Cancer.” Journal of Clinical Oncology 27(2):193 Everolimus, in Patients With Recurrent Endometrial Carcinoma.” 198. Cancer 116:5415-5419. Yao, J.C. etal. (Feb. 10, 2011). “Everolimus for Advanced Pancreatic Smith, S.M. et al. (Nov. 1, 2010, e-pub. Sep. 13, 2010). Neuroendocrine Tumors.” New England Journal of Medicine “Termsirolimus Has Activity in Non-Mantle Cell Non-Hodgkin's 364(6):514-523. Lymphoma Subtypes: The University of Chicago Phase II Consor Yao, J.C. et al. (Jan. 1, 2010, e-pub. Nov. 23, 2009). “Daily Oral tium.” Journal of Clinical Oncology 28(31):4740-4746. Everolimus Activity in Patients With Metastatic Pancreatic Soria, J-C. et al. (Oct. 2009, e-pub. Jun. 23, 2009). “Efficacy of Neuroendocrine Tumors. After Failure of Cytotoxic Chemotherapy: Everolimus (RAD001) in Patients With Advanced NSCLC Previ A Phase II Trial.” Journal of Clinical Oncology 28(1):69-76. ously Treated With Chemotherapy Alone or With Chemotherapy and Yee, K.W.L. etal. (Sep. 1, 2006). “Phase I/II Study of the Mammalian EGFR Inhibitors.” Annals of Oncology 20010): 1674-1681. Target of Rapamycin Inhibitor Everolimus (RAD001) in Patients Tabernero, J. et al. (Apr. 1, 2008, e-pub. Mar. 10, 2008). "Dose- and With Relapsed or Refractory Hematologic Malignancies.” Clin. Can Schedule-Dependent Inhibition of the Mammalian Target of cer Res. 12(17):5165-5173. Rapamycin Pathway With Everolimus: A Phase I Tumor Non-Final Office Action mailed on Feb. 13, 2014, for U.S. Appl. No. Pharmacodynamic Study in Patients With Advanced Solid Tumors.” 13/781,479, filed Feb. 28, 2013, 39 Pages. Journal of Clinical Oncology 28(10): 1603-1610. European Examination Report mailed May 9, 2014, for EP Applica Tarhini, A. etal. (Dec. 1, 2010, e-pub. Nov. 2, 2010). “Phase II Study tion No. 08767585.6, filed on May 5, 2008, 7 pages. of Everolimus (RAD001) in Previously Treated Small Cell Lung Cancer.” Clin. Cancer Res. 16(23):5900-5907. * cited by examiner U.S. Patent Dec. 16, 2014 Sheet 1 of 7 US 8,911,786 B2

U.S. Patent Dec. 16, 2014 Sheet 2 of 7 US 8,911,786 B2

6x16uuesod U.S. Patent Dec. 16, 2014 Sheet 3 of 7 US 8,911,786 B2

U.S. Patent Dec. 16, 2014 Sheet 4 of 7 US 8,911,786 B2

Figure 4

sel Safine

A -A-ABXV-Rapamycin (10mg/kg, (40mg/kg, cdb5, ip) 3xway?4uk M -g) Rapamycin + APX U.S. Patent US 8,911,786 B2

so Co so as so 009 N 000Z 009!, 000, (cuu) aunow Jouni 6ZLH U.S. Patent US 8,911,786 B2 U.S. Patent Dec. 16, 2014 Sheet 7 Of 7 US 8,911,786 B2

Figure 7.

Antitumor Activity of Nab-Rapamycin in MM1 S Multiple Myeloma Xenograft Model

7OOO -- k 6000 --

5OOO -- a -- Sol. NaC O. 9% r 4000 it -- NAB-Rapamycin 20 mg/kg i as 3x weekly 's 3000 -A- NAB-Rapamycin 40 mg/kg s 3x weekly so-NAB-Rapamycin 30 mg/kg 2OOO dailya

OOO

3 6 8 10 13 15 17 20 22 24 27 29 Days of treatment US 8,911,786 B2 1. 2 NANOPARTICLE COMPRISING RAPAMYCIN tissue. However, it is difficult (if not impossible) to selectively AND ALBUMNASANTICANCERAGENT administer therapeutic radiation to the abnormal tissue. Thus, normal tissue proximate to the abnormal tissue is also CROSS-REFERENCE TO RELATED exposed to potentially damaging doses of radiation through APPLICATIONS out the course of treatment. The efficacy of radiotherapeutic techniques in destroying abnormal proliferative cells is there This application is a National Phase filing under 35 U.S.C. fore balanced by associated cytotoxic effects on nearby nor S371 of International Application No. PCT/US2008/003096 mal cells. Because of this, radiotherapy techniques have an having an international filing date of Mar. 7, 2008, which inherently narrow therapeutic index which results in the inad claims priority benefit to provisional application Ser. No. 10 equate treatment of most tumors. Even the best radiothera 60/905,669, filed Mar. 7, 2007, provisional application Ser. peutic techniques may result in incomplete tumor reduction, No. 60/905,734, filed Mar. 7, 2007, provisional application tumor recurrence, increased tumor burden, and induction of Ser. No. 60/905,662, filed Mar. 7, 2007, provisional applica radiation resistant tumors. tion Ser. No. 60/905,735, filed Mar. 7, 2007, provisional Chemotherapy involves the disruption of cell replication or application Ser. No. 60/905,672, filed Mar. 7, 2007, provi 15 cell metabolism. Chemotherapy can be effective, but there are sional application Ser. No. 60/905,787, filed Mar. 7, 2007, severe side effects, e.g., vomiting, low white blood cells, loss provisional application Ser. No. 60/905,663, filed Mar. 7, of hair, loss of weight and other toxic effects. Because of the 2007, provisional application Ser. No. 60/905,767, filed Mar. extremely toxic side effects, many cancer individuals cannot 7, 2007, provisional application Ser. No. 60/905,750, filed Successfully finish a complete chemotherapy regime. Che Mar. 7, 2007, provisional application Ser. No. 60/923,248, motherapy-induced side effects significantly impact the qual filed Apr. 13, 2007, and provisional application Ser. No. ity of life of the individual and may dramatically influence 60/923,456, filed Apr. 13, 2007, the entire disclosures of each individual compliance with treatment. Additionally, adverse of which are hereby incorporated by reference. side effects associated with chemotherapeutic agents are gen erally the major dose-limiting toxicity in the administration TECHNICAL FIELD 25 of these drugs. For example, mucositis is one of the major dose limiting toxicities for several anticancer agents, includ This application relates to methods and compositions for ing 5-FU, , and antitumor antibiotics, such as treating, stabilizing, preventing, and/or delaying cancer using . Many of these chemotherapy-induced side nanoparticles that comprise rapamycin or a derivative thereof effects if severe may lead to hospitalization, or require treat The application further provides combination therapy meth 30 ment with analgesics for the treatment of pain. Some cancer ods of treating cancer comprising administering to an indi individuals die from the chemotherapy due to poor tolerance vidual an effective amount of nanoparticles that comprise to the chemotherapy. The extreme side effects of anticancer rapamycin or a derivative thereof and a second therapy. drugs are caused by the poor target specificity of Such drugs. The drugs circulate through most normal organs of individu BACKGROUND 35 als as well as intended target tumors. The poor target speci ficity that causes side effects also decreases the efficacy of The failure of a significant number of tumors to respond to chemotherapy because only a fraction of the drugs is correctly drug and/or radiation therapy is a serious problem in the targeted. The efficacy of chemotherapy is further decreased treatment of cancer. In fact, this is one of the main reasons by poor retention of the anti-cancer drugs within the target why many of the most prevalent forms of human cancer still 40 tumors. resist effective chemotherapeutic intervention, despite cer Another problem associated with chemotherapy is the tain advances in the field of chemotherapy. development of drug resistance. Drug resistance is the name Cancer is now primarily treated with one or a combination given to the circumstances whena disease does not respond to of three types of therapies: Surgery, radiation, and chemo a treatment drug or drugs. Drug resistance can be either therapy. Surgery is a traditional approach in which all or part 45 intrinsic, which means that disease has never been responsive of a tumor is removed from the body. Surgery generally is to the drug or drugs, or it can be acquired, which means the only effective for treating the earlier stages of cancer. While disease ceases responding to a drug or drugs that the disease Surgery is sometimes effective in removing tumors located at had previously been responsive. Combination therapy, certain sites, for example, in the breast, colon, and skin, it including combination chemotherapy, has the potential cannot be used in the treatment of tumors located in other 50 advantages of both avoiding the emergence of resistant cells areas, inaccessible to Surgeons, nor in the treatment of dis and to kill pre-existing cells which are already drug resistant. seminated neoplastic conditions such as leukemia. For more Due to the limitations of current treatments for cancer, the than 50% of cancer individuals, by the time they are diag severity and breadth of neoplasm, tumor and cancer, there nosed they are no longer candidates for effective Surgical remains a significant interest in and need for additional or treatment. Surgical procedures may increase tumor 55 alternative therapies for treating, stabilizing, preventing, and/ metastases through blood circulation during Surgery. Most of or delaying cancer. Preferably, the treatments overcome the cancer individuals do not die from the cancer at the time of shortcomings of current Surgical, chemotherapy, and radia diagnosis or Surgery, but rather die from the metastasis and tion treatments. the recurrence of the cancer. Other therapies are also often ineffective. Radiation 60 BRIEF SUMMARY OF THE INVENTION therapy is only effective for individuals who present with clinically localized disease at early and middle stages of The present invention provides methods for the treatment cancer, and is not effective for the late stages of cancer with of cancer using nanoparticles that comprise rapamycin or a metastasis. Radiation is generally applied to a defined area of derivative thereof Accordingly, the invention in some the subject’s body which contains abnormal proliferative tis 65 embodiments provides a method of treating cancer in an sue, in order to maximize the dose absorbed by the abnormal individual by administering to the individual (e.g., a human) tissue and minimize the dose absorbed by the nearby normal an effective amount of a composition comprising nanopar US 8,911,786 B2 3 4 ticles that comprise rapamycin or a derivative thereof and a not colon cancer (i.e., other than colon cancer). In some carrier protein. In some embodiments, the cancer is early embodiments, the cancer is not breast cancer (i.e., other than stage cancer, non-metastatic cancer, primary cancer, breast cancer). In some embodiments, the cancer is not ova advanced cancer, locally advanced cancer, metastatic cancer, rian cancer, prostate cancer, or brain cancer. In some embodi cancer in remission, recurrent cancer, cancer in an adjuvant ments, one or more symptoms of the cancer are ameliorated. setting, cancer in a neoadjuvant setting, or cancer Substan In some embodiments, the cancer is delayed or prevented. tially refractory to hormone therapy. In some embodiments, In some embodiments, the amount of the rapamycin or the cancer is a Solid tumor. In some embodiments, the cancer derivatives thereof in the composition (e.g., a dose or a unit is not a solid tumor (i.e., other than a solid tumor). In some dosage form) is in the range of about 54 mg to about 540 mg. embodiments, the cancer is a plasmacytoma. In some 10 such as about 180 mg to about 270 mg. or about 216 mg. In embodiments, the cancer is multiple myeloma, renal cell some embodiments, the carrier is suitable for parenteral carcinoma, prostate cancer, lung cancer, melanoma, brain administration (e.g., intravenous administration). In some cancer (e.g., glioblastoma), ovarian cancer, or breast cancer. embodiments, a is not contained in the composition. In In some embodiments, the cancer is not a carcinoma (i.e., some embodiments, the rapamycin orderivative thereof is the other than a carcinoma). In some embodiments, the cancer is 15 only pharmaceutically active agent for the treatment of cancer not colon cancer (i.e., other than colon cancer). In some that is contained in the composition. In some embodiments, embodiments, the cancer is not breast cancer (i.e., other than the composition comprises rapamycin. In some embodi breast cancer). In some embodiments, the cancer is not ova ments, the composition comprises more than about 50% of rian cancer, prostate cancer, or brain cancer. In some embodi the rapamycin or derivative thereof in nanoparticle form. In ments, one or more symptoms of the cancer are ameliorated. Some embodiments, the carrier protein is albumin, Such as In some embodiments, the cancer is delayed or prevented. human serum albumin. In some embodiments, the average In some embodiments, the amount of the rapamycin or diameter of the nanoparticles in the composition is no greater derivative thereof in the effective amount of the composition than about 200 nm (such as no greater than about 100 nm). In is in the range of about 54 mg to about 540 mg. Such as about Some embodiments, the nanoparticle compositions are sterile 180 mg to about 270 mg or about 216 mg. In some embodi 25 filterable. In some embodiments, the weight ratio of the car ments, the rapamycin or derivative thereof is administered rier protein to the rapamycin or derivative thereof in the parenterally (e.g., intravenously). In some embodiments, a nanoparticles is less than about 18:1. In some embodiments, taxane is not administered to the individual (i.e., other than a the weight ratio of the carrier protein to the rapamycin or taxane). In some embodiments, the taxane administered is not derivative thereof in the nanoparticle compositions is less a nanoparticle taxane composition. In some embodiments, 30 than about 18:1. the rapamycin or derivative thereof is the only pharmaceuti In yet another aspect, the invention includes a kit with (i) a cally active agent for the treatment of cancer that is adminis composition comprising nanoparticles that comprise rapa tered to the individual. In some embodiments, rapamycin is mycin or a derivative thereof and a carrier protein and (ii) administered. In some embodiments, the composition com instructions for use in treating cancer. In various embodi prises more than about 50% of the rapamycin or derivative 35 ments, the cancer is early stage cancer, non-metastatic cancer, thereof in nanoparticle form. In some embodiments, the car primary cancer, advanced cancer, locally advanced cancer, rier protein is albumin, such as human serum albumin. In metastatic cancer, cancer in remission, recurrent cancer, can Some embodiments, the average diameter of the nanoparticles cer in an adjuvant setting, cancer in a neoadjuvant setting, or in the composition is no greater than about 200 nm (such as no cancer Substantially refractory to hormone therapy. In some greater than about 100 nm). In some embodiments, the nano 40 embodiments, the cancer is a solid tumor. In some embodi particle compositions are sterile filterable. In some embodi ments, the cancer is a plasmacytoma. In some embodiments, ments, the weight ratio of the carrier protein to the rapamycin the cancer is multiple myeloma, renal cell carcinoma, pros or derivative thereof in the nanoparticles is less than about tate cancer, lung cancer, melanoma, brain cancer (e.g. glio 18:1. In some embodiments, the weight ratio of the carrier blastoma), ovarian cancer, or breast cancer. In some embodi protein to the rapamycin or derivative thereof in the nanopar 45 ments, the cancer is not colon cancer. In some embodiments, ticle compositions is less than about 18:1. the cancer is not breast cancer. In some embodiments, one or The invention also provides pharmaceutical compositions more symptoms of the cancer are ameliorated. In some Such as unit dosage forms that are useful for treating cancer. embodiments, the cancer is delayed or prevented. Accordingly, the invention in some embodiments provides a In some embodiments, the amount of the rapamycin or pharmaceutical composition (e.g., a unit dosage form of a 50 derivative thereof in the kit is in the range of about 54 mg to pharmaceutical composition) that includes nanoparticles that about 540 mg, such as about 180 mg to about 270 mg or about comprise rapamycin or a derivative thereof and a carrier pro 216 mg. In some embodiments, the rapamycin or derivative tein. In some embodiments, the composition also includes a thereof is administered parenterally (e.g., intravenously). In pharmaceutically acceptable carrier. In various embodi Some embodiments, the kit does not contain a taxane. In some ments, the cancer is early stage cancer, non-metastatic cancer, 55 embodiments, the rapamycin orderivative thereof is the only primary cancer, advanced cancer, locally advanced cancer, pharmaceutically active agent for the treatment of cancer that metastatic cancer, cancer in remission, recurrent cancer, can is contained in the kit. In some embodiments, the kit com cer in an adjuvant setting, cancer in a neoadjuvant setting, or prises another pharmaceutically active agent for the treatment cancer Substantially refractory to hormone therapy. In some of cancer. In some embodiments, the other pharmaceutically embodiments, the cancer is a solid tumor. In some embodi 60 active agent is a chemotherapeutic agent. In some embodi ments, the cancer is not a solid tumor (i.e., other than a Solid ments, the kit comprises rapamycin. In some embodiments, tumor). In some embodiments, the cancer is a plasmacytoma. the composition comprises more than about 50% of the rapa In some embodiments, the cancer is multiple myeloma, renal mycin or derivative thereof in nanoparticle form. In some cell carcinoma, prostate cancer, lung cancer, melanoma, brain embodiments, the carrier protein is albumin, Such as human cancer (e.g., glioblastoma), ovarian cancer, or breast cancer. 65 serum albumin. In some embodiments, the average diameter In some embodiments, the cancer is not a carcinoma (i.e., of the nanoparticles in the composition is no greater than other than a carcinoma). In some embodiments, the cancer is about 200 nm (such as no greater than about 100 nm). In some US 8,911,786 B2 5 6 embodiments, the nanoparticle compositions are sterile fil and a carrier protein and the chemotherapeutic agent are terable. In some embodiments, the weight ratio of the carrier non-concurrent. For example, in some embodiments, the protein to the rapamycin or derivative thereof in the nanopar administration of the nanoparticle composition comprising a ticles is less than about 18:1. In some embodiments, the rapamycin or a derivative thereof and a carrier protein is weight ratio of the carrier protein to the rapamycin orderiva terminated before the chemotherapeutic agent is adminis tive thereof in the nanoparticle compositions is less than tered. In some embodiments, the administration of the che about 18:1. motherapeutic agent is terminated before the nanoparticle The present invention also provides methods for the treat composition comprising a rapamycin or a derivative thereof ment of cancer using combination therapies. The invention and a carrier protein is administered. provides a method of treating cancer comprising a) a first 10 In some embodiments, there is provided a method of treat therapy comprising administering to an individual an effec ing cancer in an individual comprisinga) a first therapy com tive amount of a composition comprising nanoparticles com prising administering to the individual a composition com prising rapamycin or a derivative thereofanda carrier protein prising nanoparticles comprising rapamycin or a derivative and b) a second therapy. Such as Surgery, radiation, gene thereof and a carrier protein, and b) a second therapy com therapy, immunotherapy, bone marrow transplantation, stem 15 prising Surgery, radiation, gene therapy, immunotherapy, cell transplantation, hormonetherapy, targeted therapy, cryo bone marrow transplantation, stem cell transplantation, hor therapy, ultrasound therapy, , and/or mone therapy, targeted therapy, cryotherapy, ultrasound chemotherapy (e.g., one or more compounds or pharmaceu therapy, photodynamic therapy, or combinations thereof In tically acceptable salts thereofuseful for treating cancer). Some embodiments, the second therapy is hormone therapy. In some embodiments, the invention provides a method of In some embodiments, the second therapy is radiation treating cancer in an individual comprising administering to therapy. In some embodiments, the second therapy is Surgery. the individuala) an effective amount of a composition com In some embodiments, the first therapy is carried out prior to prising nanoparticles comprising a rapamycin or a derivative the second therapy. In some embodiments, the first therapy is thereof and a carrier protein and b) an effective amount of at carried out after the second therapy. least one other chemotherapeutic agent. In some embodi 25 In some embodiments, the cancer being treated by combi ments, the chemotherapeutic agent is any of (and in some nation therapy is early stage cancer, non-metastatic cancer, embodiments selected from the group consisting of) taxane, primary cancer, advanced cancer, locally advanced cancer, (including nucleoside analogs), platinum metastatic cancer, cancer in remission, recurrent cancer, can based agents, alkylating agents, tyrosine kinase inhibitors, cer in an adjuvant setting, cancer in a neoadjuvant setting, or antibiotics, Vinca alkloids, proteasome inhibi 30 cancer Substantially refractory to hormone therapy. In some tors, modulators of HER2/neu, modulators of EGFR, modu embodiments, the cancer is a solid tumor. In some embodi lators of VEGFR, and topoisomerase inhibitors. In some ments, the cancer is not a solid tumor (i.e., other than a solid embodiments, the chemotherapeutic agent is a platinum tumor). In some embodiments, the cancer is a plasmacytoma. based agent, such as carboplatin. In some embodiments, the In some embodiments, the cancer is multiple myeloma, renal chemotherapeutic agent is a modulator of HER2/neu (Such as 35 cell carcinoma, prostate cancer, lung cancer, melanoma, brain an inhibitor of HER2/neu for example HerceptinR). In some cancer (e.g., glioblastoma), ovarian cancer, or breast cancer. embodiments, the chemotherapeutic agent is a modulator of In some embodiments, the cancer is not a carcinoma (i.e., EGFR (such as an inhibitor of EGFR for example Erbitux(R). other than a carcinoma). In some embodiments, the cancer is In some embodiments, the chemotherapeutic agent is an anti not colon cancer (i.e., other than colon cancer). In some VEGF antibody (such as bevacizumab, e.g., Avastin R). In 40 embodiments, the cancer is not breast cancer (i.e., other than Some embodiments, the effective amounts of the nanoparticle breast cancer). In some embodiments, the cancer is not ova composition and the anti-VEGF antibody synergistically rian cancer, prostate cancer, or brain cancer. In some embodi inhibit cell proliferation or metastasis. In some embodiments, ments, one or more symptoms of the cancer are ameliorated. the chemotherapeutic agent affects a signaling pathway In some embodiments, the cancer is delayed or prevented. involving a target of rapamycin. In some embodiments the 45 In some embodiments, the amount of the rapamycin or chemotherapeutic agent affects a signaling pathway involv derivative thereof in the effective amount of the composition ing mTOR (such as the PI3K/Akt signaling pathway). In used in combination therapy is in the range of about 54 mg to Some embodiments, a taxane is not administered to the indi about 540 mg, such as about 180 mg to about 270 mg or about vidual. In some embodiments, the taxane administered is not 216 mg. In some embodiments, the rapamycin or derivative in a nanoparticle composition. 50 thereof is administered parenterally (e.g., intravenously). In In some embodiments, the composition comprising nano Some embodiments, a taxane is not administered to the indi particles comprising a rapamycin or a derivative thereof and vidual (i.e., other than taxane). In some embodiments, the a carrier protein and the chemotherapeutic agent are admin taxane administered is not a nanoparticle taxane composition. istered simultaneously, either in the same composition or in In some embodiments, rapamycin is administered. In some separate compositions. In some embodiments, the nanopar 55 embodiments, the composition comprises more than about ticle composition comprising a rapamycin or a derivative 50% of the rapamycin or derivative thereof in nanoparticle thereof and a carrier protein and the chemotherapeutic agent form. In some embodiments, the carrier protein is albumin, are administered sequentially, e.g., the nanoparticle compo Such as human serum albumin. In some embodiments, the sition is administered either prior to or after the administra average diameter of the nanoparticles in the composition is no tion of the chemotherapeutic agent. In some embodiments, 60 greater than about 200 nm (such as no greater than about 100 the administration of the nanoparticle composition compris nm). In some embodiments, the nanoparticle compositions ing a rapamycin or a derivative thereof and a carrier protein are sterile filterable. In some embodiments, the weight ratio and the chemotherapeutic agent are concurrent, e.g., the of the carrier protein to the rapamycin orderivative thereof in administration period of the nanoparticle composition and the nanoparticles is less than about 18:1. In some embodi that of the chemotherapeutic agent overlap with each other. In 65 ments, the weight ratio of the carrier protein to the rapamycin Some embodiments, the administration of the nanoparticle or derivative thereof in the nanoparticle compositions is less composition comprising a rapamycin or a derivative thereof than about 18:1. US 8,911,786 B2 7 8 The invention also provides pharmaceutical compositions nation therapy context. For example, a kit may provide Such Such as unit dosage forms that are useful in combination a composition in addition to another therapeutic composition. therapy for treating cancer. Accordingly, the invention in In some embodiments, the instructions are instructions for Some embodiments provides a pharmaceutical composition providing a first and second therapy, wherein either the first or (e.g., a unit dosage form of a pharmaceutical composition) for second therapy comprises administering a composition that use in combination therapy that includes nanoparticles that comprises nanoparticles of rapamycin or derivative thereof comprise rapamycin or a derivative thereof and a carrier pro and a carrier protein. In some embodiments, the kit further tein. In some embodiments, the pharmaceutical composition comprises at least one other therapeutic agent. In some includes a) nanoparticles comprising rapamycin or a deriva embodiments, the other therapeutic agent comprises a che tive thereof and a carrier protein and b) at least one other 10 motherapeutic agent. In some embodiments, the other thera therapeutic agent. In some embodiments, the other therapeu peutic agent comprises a hormonetherapeutic agent. In some tic agent comprises a chemotherapeutic agent. In some embodiments, the cancer is early stage cancer, non-metastatic embodiments, the other therapeutic agent comprises a hor cancer, primary cancer, advanced cancer, locally advanced mone therapeutic agent. In some embodiments, the compo cancer, metastatic cancer, cancer in remission, recurrent can sition also includes a pharmaceutically acceptable carrier. In 15 cer, cancer in an adjuvant setting, cancer in a neoadjuvant Some embodiments, the cancer is early stage cancer, non setting, or cancer Substantially refractory to hormonetherapy. metastatic cancer, primary cancer, advanced cancer, locally In some embodiments, the cancer is a Solid tumor. In some advanced cancer, metastatic cancer, cancer in remission, embodiments, the cancer is not a solid tumor (i.e., other than recurrent cancer, cancer in an adjuvant setting, cancer in a a solid tumor). In some embodiments, the cancer is a plasma neoadjuvant setting, or cancer Substantially refractory to hor cytoma. In some embodiments, the cancer is multiple mone therapy. In some embodiments, the cancer is a Solid myeloma, renal cell carcinoma, prostate cancer, lung cancer, tumor. In some embodiments, the cancer is not a solid tumor melanoma, brain cancer (e.g., glioblastoma), ovarian cancer, (i.e., other than a Solid tumor). In some embodiments, the or breast cancer. In some embodiments, the cancer is a carci cancer is a plasmacytoma. In some embodiments, the cancer noma (i.e., other than a carcinoma). In some embodiments, is multiple myeloma, renal cell carcinoma, prostate cancer, 25 the cancer is not colon cancer (i.e., other than colon cancer). lung cancer, melanoma, brain cancer (e.g., glioblastoma). In some embodiments, the cancer is not breast cancer (i.e., ovarian cancer, or breast cancer. In some embodiments, the other than breast cancer). In some embodiments, the cancer is cancer is a carcinoma (i.e., other than a carcinoma). In some not ovarian cancer, prostate cancer, or brain cancer. In some embodiments, the cancer is not colon cancer (i.e., other than embodiments, one or more symptoms of the cancer are ame colon cancer). In some embodiments, the cancer is not breast 30 liorated. In some embodiments, the cancer is delayed or pre cancer (i.e., other than breast cancer). In some embodiments, vented. the cancer is not ovarian cancer, prostate cancer, or brain In some embodiments, the amount of the rapamycin or cancer. In some embodiments, one or more symptoms of the derivative thereof in the kit for use in combination therapy is cancer are ameliorated. In some embodiments, the cancer is in the range of about 54 mg to about 540 mg. Such as about delayed or prevented. 35 180 mg to about 270 mg or about 216 mg. In some embodi In some embodiments, the amount of the rapamycin or ments, the rapamycin or derivative thereof is administered derivatives thereof in the composition (e.g., a dose or a unit parenterally (e.g., intravenously). In some embodiments, the dosage form) for use in combination therapy is in the range of kit does not contain a taxane. In some embodiments, the about 54 mg to about 540 mg, such as about 180 mg to about rapamycin or derivative thereof is the only pharmaceutically 270 mg, or about 216 mg. In some embodiments, the carrier 40 active agent for the treatment of cancer that is contained in the is suitable for parenteral administration (e.g., intravenous kit. In some embodiments, the kit comprises another pharma administration). In some embodiments, a taxane is not con ceutically active agent for the treatment of cancer. In some tained in the composition. In some embodiments, the rapa embodiments, the other pharmaceutically active agent is a mycin or derivative thereof is the only pharmaceutically chemotherapeutic agent. In some embodiments, the kitcom active agent for the treatment of cancer that is contained in the 45 prises rapamycin. In some embodiments, the composition composition (for example, as part of a kit that contains comprises more than about 50% of the rapamycin or deriva instructions for using the composition with another therapy). tive thereof in nanoparticle form. In some embodiments, the In some embodiments, the composition comprises rapa carrier protein is albumin, Such as human serum albumin. In mycin. In some embodiments, the composition comprises Some embodiments, the average diameter of the nanoparticles more than about 50% of the rapamycin orderivative thereof in 50 in the composition is no greater than about 200 nm (such as no nanoparticle form. In some embodiments, the carrier protein greater than about 100 nm). In some embodiments, the nano is albumin, Such as human serum albumin. In some embodi particle compositions are sterile filterable. In some embodi ments, the average diameter of the nanoparticles in the com ments, the weight ratio of the carrier protein to the rapamycin position is no greater than about 200 nm (such as no greater or derivative thereof in the nanoparticles is less than about than about 100 nm). In some embodiments, the nanoparticle 55 18:1. In some embodiments, the weight ratio of the carrier compositions are sterile filterable. In some embodiments, the protein to the rapamycin or derivative thereof in the nanopar weight ratio of the carrier protein to the rapamycin orderiva ticle compositions is less than about 18:1. tive thereof in the nanoparticles is less than about 18:1. In The invention also provides any of the compositions Some embodiments, the weight ratio of the carrier protein to described (e.g., a composition comprising nanoparticles that the rapamycin or derivative thereof in the nanoparticle com 60 comprise rapamycin or a derivative thereof and a carrier pro positions is less than about 18:1. tein) for any use described herein whether in the context of In yet another aspect, the invention includes a kit with (i) a use as a medicament and/or use for manufacture of a medi composition comprising nanoparticles that comprise rapa cament. Also provided are unit dosage forms of compositions mycin or a derivative thereof and a carrier protein and (ii) described herein, articles of manufacture comprising the instructions for use in combination therapy for treating can 65 inventive compositions or unit dosage forms in Suitable pack cer. The invention also provides kits for using the rapamycin aging (e.g., vials or vessels including sealed vials or vessels (or its derivatives) compositions described herein in combi and sterile sealed vials or vessels), and kits comprising the US 8,911,786 B2 10 unit dosage forms. The invention also provides methods of upregulated in Some cancer cells (e.g., breast cancer cells). making and using these compositions as described herein. This increased specificity of Nab-rapamycin may increase the It is to be understood that one, some, or all of the properties effectiveness of rapamycin and may allow lower doses of of the various embodiments described herein may be com rapamycin to be used, which would minimize toxic effects bined to form other embodiments of the present invention. from rapamycin while still inhibiting, stabilizing, preventing, or delaying tumor growth. The increased specificity may also BRIEF DESCRIPTION OF FIGURES reduce toxic side-effects from interactions of rapamycin with noncancerous cells and tissues, such as intestinal toxicity that FIG. 1 is a table listing the intravenous pharmacokinetic Sometimes limits the dose of rapamycin that can be given to a parameters for the albumin-containing nanoparticle formula 10 patient. The nanoparticle formulation of rapamycin also tion of rapamycin (hereinafter referred to as Nab-rapamycin). increases the Solubility of rapamycin and allows larger doses FIG. 2A is a graph of Cmax versus dose, showing linearity to be used, if desired. for the Nab-rapamycin. Definitions FIG. 2B is a graph of AUC versus dose, showing linearity As used herein, “the composition” or “compositions' for Nab-rapamycin. 15 includes and is applicable to compositions of the invention. FIG. 2C is a graph of Vss versus dose, showing possible The invention also provides pharmaceutical compositions saturable volume of distribution for Nab-rapamycin. comprising the components described herein. FIG. 2D is a graph showing the log-linear plot of Nab The term, “rapamycin herein refers to rapamycin or its rapamycin blood concentration vs. time following IV admin derivatives and accordingly the invention contemplates and istration to rats at dose levels of 15 mg/kg, 30 mg/kg, and 45 includes all these embodiments. Rapamycin is sometimes mg/kg. referred to elsewhere as sirolimus, rapammune, or rapamune. FIG. 3A is a graph of the antitumor activity of Nab-rapa Reference to “rapamycin' is to simplify the description and is mycin in mice with MX-1 breast tumor xenografts. exemplary. Derivatives of rapamycin include, but are not FIG. 3B is a graph of the weight loss in mice with MX-1 limited to, compounds that are structurally similar to rapamy breast tumor xenografts after the administration of Nab-rapa 25 cin, or are in the same general chemical class as rapamycin, mycin or saline. analogs of rapamycin, orpharmaceutically acceptable salts of FIG. 4 is a graph showing the antitumor activity of Abrax rapamycin or its derivatives or analogs. In some embodi aneTM, Nab-rapamycin, and Nab-rapamycin in combination ments, rapamycin or a derivative thereof increases basal AKT with AbraxaneTM in mice with HT29 colon tumor xenografts. activity, increases AKT phosphorylation, increases PI3-ki FIG. 5A is a graph showing the antitumor activity of Nab 30 nase activity, increases the length of activation of AKT (e.g., rapamycin in mice with HT29 colon tumor Xenografts. activation induced by exogenous IGF-1), inhibits serine FIG. 5B is a graph showing the weight loss in mice with phosphorylation of IRS-1, inhibits IRS-1 degradation, inhib H29 colon tumor xenografts after the administration of Nab its or alters CXCR4 subcellular localization, inhibits VEGF rapamycin or DMSO. secretion, decreases expression of cyclin D2, decreases FIG. 6A is a graph showing the antitumor activity of Nab 35 expression of survivin, inhibits IL-6-induced multiple rapamycin in mice with HCT-116 colon tumor xenografts. myeloma cell growth, inhibits cancer cell proliferation, FIG. 6B is a graph showing the weight loss in mice with increases apoptosis, increases arrest, increases HCT-116 colon tumor xenografts after the administration of cleavage of poly(ADPribose) polymerase, increases cleavage Nab-rapamycin or saline. of caspase-8/caspase-9, alters or inhibits signaling in the FIG. 7 is a graph showing the antitumor activity of Nab 40 phosphatidylinositol 3-kinase/AKT/mTOR and/or cyclin rapamycin in mice with MM1S multiple myeloma tumor D1/retinoblastoma pathways, inhibits angiogenesis, and/or Xenografts. inhibits osteoclast formation. In some embodiments, the derivative of rapamycin retains one or more similar biologi DETAILED DESCRIPTION OF THE INVENTION cal, pharmacological, chemical and/or physical properties 45 (including, for example, functionality) as rapamycin. In some The present invention provides methods, compositions, embodiments, the rapamycin derivative has at least about any and kits for the treatment or prevention of cancer using nano of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or particles that comprise rapamycin or a derivative thereof and 100% of an activity of rapamycin. For example, the decrease a carrier protein (Such as albumin). The present invention in the size of a tumor, the number of cancer cells, or the further provides methods, compositions, and kits for use in 50 growth rate of a tumor caused by a rapamycin derivative is combination therapy for the treatment or prevention of cancer preferably at least about any of 10%, 20%, 30%, 40%, 50%, using nanoparticles that comprise rapamycin or a derivative 60%, 70%, 80%, 90%. 95% or 100% of the corresponding thereof and a carrier protein (such as albumin). Any of these decrease caused by the same amount of rapamycin. An exem compositions can be used to treat, stabilize, prevent, and/or plary rapamycin derivative includes benzoyl rapamycin, Such delay cancer. 55 as that disclosed in paragraph (0022 of WO 2006/089207, In particular, nanoparticles comprising rapamycin (also which is hereby incorporated by reference in its entirety. referred to as a "nanoparticle composition') and the carrier Other exemplary rapamycin derivatives include WY-090217. protein albumin were shown to significantly inhibit the AY-22989, NSC-226080, SiiA-9268A, oxaazacyclohentria growth of a human mammary carcinoma implanted into a contine, temsirolimus (CCI 779 (Wyeth)), everolimus (RAD mouse model (Example 3) and inhibit tumor growth in mice 60 001 (Novartis)), pimecrolimus (ASM981), SDZ-RAD, with MM1S multiple myeloma tumor xenografts (Example SAR943, ABT-578, AP23573, and Biolimus A9. 12B). This albumin-containing nanoparticle formulation of Unless clearly indicated otherwise, “an individual' as used rapamycin was nontoxic at the doses tested and displayed herein intends a mammal, including but not limited to a pri linear pharmacokinetics with respect to dose (Example 2). mate, human, bovine, horse, feline, canine, or rodent. The nanoparticle formulation of albumin and rapamycin 65 As used herein, “treatment' or “treating is an approach for enhances tumor penetration through albumin receptor obtaining beneficial or desired results including clinical (gp60)-mediated binding of the SPARC protein, which is results. For purposes of this invention, beneficial or desired US 8,911,786 B2 11 12 clinical results include, but are not limited to, one or more of As used herein, by “combination therapy” is meant a first the following: decreasing one more symptoms resulting from therapy that includes nanoparticles comprising rapamycin or the disease, diminishing the extent of the disease, stabilizing a derivative thereof and a carrier protein in conjunction with the disease (e.g., preventing or delaying the worsening of the a second therapy (e.g., radiation, Surgery, or chemotherapeu disease), preventing or delaying the spread (e.g., metastasis) tic agent) useful for treating, stabilizing, preventing, and/or of the disease, preventing or delaying the occurrence or recur delaying cancer. Administration in "conjunction with rence of the disease, delay or slowing the progression of the another compound includes administration in the same or disease, ameliorating the disease state, providing a remission different composition(s), either sequentially, simultaneously, (whether partial or total) of the disease, decreasing the dose of or continuously. In some variations, the combination therapy one or more other required to treat the disease, 10 optionally includes one or more pharmaceutically acceptable delaying the progression of the disease, increasing the quality carriers or excipients, non-pharmaceutically active com of life, and/or prolonging Survival. In some embodiments, the pounds, and/or inert Substances. composition reduces the severity of one or more symptoms The term “effective amount' intends such amount of a associated with cancer by at least about any of 10%. 20%, composition (e.g., nanoparticles that comprise rapamycin or 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100% com 15 a derivative thereofanda carrier protein), first therapy, second pared to the corresponding symptom in the same subject prior therapy, or a combination therapy, which in combination with to treatment or compared to the corresponding symptom in its parameters of efficacy and toxicity, should be effective in other subjects not receiving the composition. Also encom a given therapeutic form based on the knowledge of the prac passed by “treatment' is a reduction of pathological conse ticing specialist. In various embodiments, an effective quence of cancer. The methods of the invention contemplate amount of the composition or therapy may (i) reduce the any one or more of these aspects of treatment. number of cancer cells; (ii) reduce tumor size; (iii) inhibit, As used herein, “delaying the development of cancer retard, slow to some extent, and preferably stop cancer cell means to defer, hinder, slow, retard, stabilize, and/or postpone infiltration into peripheral organs; (iv) inhibit (e.g., slow to development of the disease. This delay can be of varying Some extent and preferably stop) tumor metastasis; (V) inhibit lengths of time, depending on the history of the disease and/or 25 tumor growth; (vi) prevent or delay occurrence and/or recur individual being treated. As is evident to one skilled in the art, rence of a tumor, and/or (vii) relieve to some extent one or a Sufficient or significant delay can, in effect, encompass more of the symptoms associated with the cancer. In various prevention, in that the individual does not develop the disease. embodiments, the amount is Sufficient to ameliorate, palliate, A method that “delays’ development of cancer is a method lessen, and/or delay one or more of symptoms of cancer. that reduces probability of disease development in a given 30 In some embodiments, the amount of the composition, first time frame and/or reduces the extent of the disease in a given therapy, second therapy, or combination therapy is an amount time frame, when compared to not using the method. Such sufficient to decrease the size of a tumor, decrease the number comparisons are typically based on clinical studies, using a of cancer cells, or decrease the growth rate of a tumor by at statistically significant number of Subjects. Cancer develop least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, ment can be detectable using standard methods. Such as rou 35 80%, 90%, 95% or 100% compared to the corresponding tine physical exams, mammography, imaging, or biopsy. tumor size, number of cancer cells, or tumor growth rate in the Development may also refer to disease progression that may same subject prior to treatment or compared to the corre be initially undetectable and includes occurrence, recurrence, sponding activity in other Subjects not receiving the treat and onset. ment. Standard methods can be used to measure the magni As used herein, an "at risk” individual is an individual who 40 tude of this effect, such as in vitro assays with purified is at risk of developing cancer. An individual'at risk” may or enzyme, cell-based assays, animal models, or human testing. may not have detectable disease, and may or may not have As is understood in the art, an “effective amount” may be in displayed detectable disease prior to the treatment methods one or more doses, i.e., a single dose or multiple doses may be described herein. At risk” denotes that an individual has one required to achieve the desired treatment endpoint. An effec or more so-called risk factors, which are measurable param 45 tive amount may be considered in the context of administer eters that correlate with development of cancer, which are ing one or more therapeutic agents, and a nanoparticle com described herein. An individual having one or more of these position (e.g., a composition including rapamycin and a risk factors has a higher probability of developing cancer than carrier protein) may be considered to be given in an effective an individual without these risk factor(s). amount if, in conjunction with one or more other agents, a Adjuvant setting refers to a clinical setting in which an 50 desirable or beneficial result may be or is achieved. The individual has had a history of cancer, and generally (but not components (e.g., the first and second therapies) in a combi necessarily) been responsive to therapy, which includes, but is nation therapy of the invention may be administered sequen not limited to, Surgery (e.g., Surgical resection), radiotherapy, tially, simultaneously, or continuously using the same or dif and chemotherapy. However, because of their history of the ferent routes of administration for each component. Thus, an cancer, these individuals are considered at risk of develop 55 effective amount of a combination therapy includes an ment of the disease. Treatment or administration in the “adju amount of the first therapy and an amount of the second vant setting refers to a subsequent mode of treatment. The therapy that when administered sequentially, simultaneously, degree of risk (e.g., when an individual in the adjuvant setting or continuously produces a desired outcome. is considered as “high risk” or “low risk”) depends upon A “therapeutically effective amount” refers to an amount several factors, most usually the extent of disease when first 60 of a composition (e.g., nanoparticles that comprise rapamy treated. cin or a derivative thereof and a carrier protein), first therapy, “Neoadjuvant setting refers to a clinical setting in which second therapy, or a combination therapy Sufficient to pro the method is be carried out before the primary/definitive duce a desired therapeutic outcome (e.g., reducing the sever therapy. ity or duration of stabilizing the severity of, or eliminating As used herein, by “pharmaceutically active compound is 65 one or more symptoms of cancer). For therapeutic use, ben meant a chemical compound that induces a desired effect, eficial or desired results include, e.g., decreasing one or more e.g., treating, stabilizing, preventing, and/or delaying cancer. symptoms resulting from the disease (biochemical, histologic US 8,911,786 B2 13 14 and/or behavioral), including its complications and interme 10-103 colony forming units at a temperature in the range of diate pathological phenotypes presenting during develop 20 to 25°C.). Other indicia of significant microbial growth ment of the disease, increasing the quality of life of those are described in U.S. Ser. No. 11/514,030, filed Aug. 30, Suffering from the disease, decreasing the dose of other medi 2006, which is hereby incorporated by reference in its cations required to treat the disease, enhancing effect of 5 entirety. another , delaying the progression of the disease, "Sugar as used herein includes, but is not limited to, and/or prolonging Survival of patients. monosaccharides, disaccharides, polysaccharides, and A "prophylactically effective amount” refers to an amount derivatives or modifications thereof. Suitable sugars for com of a composition (e.g., nanoparticles that comprise rapamy positions described herein include, for example, mannitol, cin or a derivative thereof and a carrier protein), first therapy, 10 Sucrose, fructose, lactose, maltose, and trehalose. second therapy, or a combination therapy Sufficient to prevent As used herein, by “pharmaceutically acceptable' or or reduce the severity of one or more future symptoms of “pharmacologically compatible' is meant a material that is cancer when administered to an individual who is susceptible not biologically or otherwise undesirable, e.g., the material and/or who may develop cancer. For prophylactic use, ben may be incorporated into a pharmaceutical composition eficial or desired results include, e.g., results such as elimi 15 administered to a patient without causing any significant nating or reducing the risk, lessening the severity of future undesirable biological effects or interacting in a deleterious disease, or delaying the onset of the disease (e.g., delaying manner with any of the other components of the composition biochemical, histologic and/or behavioral symptoms of the in which it is contained. Pharmaceutically acceptable carriers disease, its complications, and intermediate pathological phe or excipients have preferably met the required standards of notypes presenting duringfuture development of the disease). toxicological and manufacturing testing and/or are included The term “simultaneous administration, as used herein, on the Inactive Ingredient Guide prepared by the U.S. Food means that a first therapy and second therapy in a combination and Drug administration. therapy are administered with a time separation of no more As used herein, reference to “not a value or parameter than about 15 minutes. Such as no more than about any of 10, generally means and describes “other than a value or param 5, or 1 minutes. When the first and second therapies are 25 eter. For example, if a taxane is not administered, it means an administered simultaneously, the first and second therapies agent other than a taxane is administered. may be contained in the same composition (e.g., a composi Reference to “about a value or parameter herein includes tion comprising both a first and second therapy) or in separate (and describes) embodiments that are directed to that value or compositions (e.g., a first therapy in one composition and a parameter perse. For example, description referring to “about second therapy is contained in another composition). 30 X” includes description of “X”. As used herein, the term “sequential administration' As used herein and in the appended claims, the singular means that the first therapy and second therapy in a combi forms “a” “or” and “the include plural referents unless the nation therapy are administered with a time separation of context clearly dictates otherwise. It is understood that aspect more than about 15 minutes, such as more than about any of and embodiments of the invention described herein include 20, 30, 40, 50, 60, or more minutes. Either the first therapy or 35 “consisting and/or "consisting essentially of aspects and the second therapy may be administered first. The first and embodiments. second therapies are contained in separate compositions, Methods of Treating Cancer which may be contained in the same or different packages or The invention provides methods of treating cancer in an kits. individual (e.g., human) comprising administering to the The term “proteins’ refers to polypeptides or polymers of 40 individual an effective amount of a composition comprising amino acids of any length (including full length or frag nanoparticles comprising rapamycin or a derivative thereof ments), which may be linear or branched, comprise modified and a carrier protein (e.g., albumin). The present invention amino acids, and/or be interrupted by non-amino acids. The provides a method of treating cancer in an individual (e.g., term also encompasses an amino acid polymer that has been human) comprising administering to the individual an effec modified naturally or by intervention, including, for example, 45 tive amount of a composition comprising nanoparticles com disulfide bond formation, glycosylation, lipidation, acetyla prising rapamycin and an albumin. The therapy may be tion, phosphorylation, or any other manipulation or modifi monotherapy or in a combination therapy context. Addition cation. Also included within this term are, for example, ally, the invention provides a method of treating cancer in an polypeptides containing one or more analogs of an amino individual by administering to the individual an effective acid (including, for example, unnatural amino acids, etc.), as 50 amount of a combination of a) a first therapy that comprises a well as other modifications known in the art. The proteins composition comprising nanoparticles that comprise rapa described herein may be naturally-occurring, i.e., obtained or mycin or a derivative thereof and a carrier protein (e.g., albu derived from a natural Source (e.g., blood) or synthesized min) and b) a second therapy useful for treating cancer. In (e.g., chemically synthesized or by synthesized by recombi Some embodiments, the second therapy includes Surgery, nant DNA techniques). Exemplary carrier proteins are 55 radiation, gene therapy, immunotherapy, bone marrow trans described herein. plantation, stem cell transplantation, hormone therapy, tar The term “antimicrobial agent used herein refers to an geted therapy, cryotherapy, ultrasound therapy, photody agent that is capable of inhibiting (e.g., delaying, reducing, namic therapy, and/or chemotherapy (e.g., one or more slowing, and/or preventing) the growth of one or more micro compounds useful for treating cancer). It is understood that organisms. Significant microbial growth can be measured or 60 reference to and description of methods of treating cancer indicated by a number of ways known in the art, Such as one below is exemplary and that this description applies equally or more of the following: (i) microbial growth in a composi to and includes methods of treating cancer using combination tion that is enough to cause one or more adverse effects to an therapy. individual when the composition is administered to the indi Examples of cancers that may be treated by the methods of vidual; (ii) more than about 10-fold increase in microbial 65 the invention include, but are not limited to, adenocortical growth over a certain period of time (for example over a 24 carcinoma, agnogenic myeloid metaplasia, AIDS-related hour period) upon extrinsic contamination (e.g., exposure to cancers (e.g., AIDS-related lymphoma), anal cancer, appen US 8,911,786 B2 15 16 dix cancer, astrocytoma (e.g., cerebellar and cerebral), basal (e.g., precursor B-lymphoblastic leukemia/lymphoma) and cell carcinoma, bile duct cancer (e.g., extrahepatic), bladder peripheral B-cell neoplasms (e.g., B-cell chronic lympho cancer, bone cancer, (osteosarcoma and malignant fibrous cytic leukemia/prolymphocytic leukemia/small lymphocytic histiocytoma), brain tumor (e.g., glioma, brain stem glioma, lymphoma (Small lymphocytic (SL) NHL), lymphoplasma cerebellar or cerebral astrocytoma (e.g., pilocytic astrocy cytoid lymphoma? immunocytoma, mantel cell lymphoma, toma, diffuse astrocytoma, anaplastic (malignant) astrocy follicle center lymphoma, follicular lymphoma (e.g., cyto toma), malignant glioma, ependymoma, oligodenglioma, logic grades: I (Small cell), II (mixed Small and large cell), III meningioma, craniopharyngioma, haemangioblastomas, (large cell) and/or subtype: diffuse and predominantly small medulloblastoma, Supratentorial primitive neuroectodermal cell type), low grade/follicular non-Hodgkin’s lymphoma tumors, visual pathway and hypothalamic glioma, and glio 10 (NHL), intermediate grade/follicular NHL, marginal Zone blastoma), breast cancer, bronchial adenomas/carcinoids, carcinoid tumor (e.g., gastrointestinal carcinoid tumor), car B-cell lymphoma (e.g., extranodal (e.g., MALT-type +/- cinoma of unknown primary, central nervous system lym monocytoid B cells) and/or Nodal (e.g., +/- monocytoid B phoma, cervical cancer, colon cancer, colorectal cancer, cells)), splenic marginal Zone lymphoma (e.g., +/- villous chronic myeloproliferative disorders, endometrial cancer 15 lymphocytes), Hairy cell leukemia, plasmacytoma/plasma (e.g., uterine cancer), ependymoma, esophageal cancer, cell myeloma (e.g., myeloma and multiple myeloma), diffuse Ewing's family of tumors, eye cancer (e.g., intraocular mela large B-cell lymphoma (e.g., primary mediastinal (thymic) noma and retinoblastoma), gallbladder cancer, gastric (stom B-cell lymphoma), intermediate grade diffuse NHL, Bur ach) cancer, gastrointestinal carcinoid tumor, gastrointestinal kitt's lymphoma, High-grade B-cell lymphoma, Burkitt-like, stromal tumor (GIST), germ cell tumor, (e.g., extracranial, high grade immunoblastic NHL, high grade lymphoblastic extragonadal, ovarian), gestational trophoblastic tumor, head NHL, high grade small non-cleaved cell NHL, bulky disease and neck cancer, hepatocellular (liver) cancer (e.g., hepatic NHL, AIDS-related lymphoma, and Waldenstrom's macro carcinoma and heptoma), hypopharyngeal cancer, islet cell globulinemia). carcinoma (endocrine pancreas), laryngeal cancer, laryngeal In some embodiments the lymphoid neoplasm (e.g., lym cancer, leukemia, lip and oral cavity cancer, oral cancer, liver 25 phoma) is a T-cell and/or putative NK-cell neoplasm. cancer, lung cancer (e.g., Small cell lung cancer, non-Small Examples of T-cell and/or putative NK-cell neoplasms cell lung cancer, adenocarcinoma of the lung, and squamous include, but are not limited to, precursor T-cell neoplasm carcinoma of the lung), lymphoid neoplasm (e.g., lym (precursor T-lymphoblastic lymphomafleukemia) and phoma), medulloblastoma, melanoma, mesothelioma, meta peripheral T-cell and NK-cell neoplasms (e.g., T-cell chronic static Squamous neck cancer, mouth cancer, multiple endo 30 lymphocytic leukemia/prolymphocytic leukemia, and large crine neoplasia syndrome, myelodysplastic syndromes, granular lymphocyte leukemia (LGL) (e.g., T-cell type and/or myelodysplastic/myeloproliferative diseases, nasal cavity NK-cell type), cutaneous T-cell lymphoma (e.g., mycosis and paranasal sinus cancer, nasopharyngeal cancer, neuro fungoides/Sezary syndrome), primary T-cell lymphomas blastoma, neuroendocrine cancer, oropharyngeal cancer, ova unspecified (e.g., cytological categories (e.g., medium-sized rian cancer (e.g., ovarian epithelial cancer, ovarian germ cell 35 cell, mixed medium and large cell), large cell, lymphoepithe tumor, ovarian low malignant potential tumor), pancreatic loid cell, Subtype hepatosplenic Yö T-cell lymphoma, and cancer, parathyroid cancer, penile cancer, cancer of the peri Subcutaneous panniculitic T-cell lymphoma), angioimmuno toneal, pharyngeal cancer, pheochromocytoma, pineoblas blastic T-cell lymphoma (AILD), angiocentric lymphoma, toma and Supratentorial primitive neuroectodermal tumors, intestinal T-cell lymphoma (e.g., +/- enteropathy associated), pituitary tumor, pleuropulmonary blastoma, lymphoma, pri 40 adult T-cell lymphomafleukemia (ATL), anaplastic large cell mary central nervous system lymphoma (microglioma), pull lymphoma (ALCL) (e.g., CD30+, T- and null-cell types), monary lymphangiomyomatosis, rectal cancer, renal cancer, anaplastic large-cell lymphoma, and Hodgkin’s like). renal pelvis and ureter cancer (transitional cell cancer), rhab In some embodiments the lymphoid neoplasm (e.g., lym domyosarcoma, salivary gland cancer, skin cancer (e.g., non phoma) is Hodgkin’s disease. For example, the Hodgkin’s melanoma (e.g., Squamous cell carcinoma), melanoma, and 45 disease may be lymphocyte predominance, nodular Sclerosis, Merkel cell carcinoma), Small intestine cancer, squamous cell mixed cellularity, lymphocyte depletion, and/or lymphocyte cancer, testicular cancer, throat cancer, thymoma and thymic rich. carcinoma, thyroid cancer, tuberous Sclerosis, urethral can In some embodiments, there are provided methods of treat cer, vaginal cancer, Vulvar cancer, Wilms tumor, and post ing cancer in an individual by administering to the individual transplant lymphoproliferative disorder (PTLD), abnormal 50 (e.g., a human) an effective amount of a composition com vascular proliferation associated with phakomatoses, edema prising nanoparticles comprising rapamycin or a derivative (such as that associated with brain tumors), and Meigs' Syn thereof and a carrier protein (e.g., albumin), wherein the drome. cancer is leukemia. In some embodiments, there are provided In some embodiments, there are provided methods of treat methods of treating cancer in an individual by administering ing cancer in an individual by administering to the individual 55 to the individual (e.g., a human) an effective amount of a (e.g., a human) an effective amount of a composition com composition comprising nanoparticles comprising a rapamy prising nanoparticles comprising rapamycin or a derivative cin and an albumin, wherein the cancer is leukemia. In some thereof and a carrier protein (e.g., albumin), wherein the embodiments, the leukemia is chronic leukemia. Examples of cancer is a lymphoid neoplasm (e.g., lymphoma). In some chronic leukemia include, but are not limited to, chronic embodiments, there are provided methods of treating cancer 60 myelocytic I (granulocytic) leukemia, chronic myelogenous, in an individual by administering to the individual (e.g., a and chronic lymphocytic leukemia (CLL). In some embodi human) an effective amount of a composition comprising ments, the leukemia is acute leukemia. Examples of acute nanoparticles comprising a rapamycin and an albumin, leukemia include, but are not limited to, acute lymphoblastic wherein the cancer is alymphoid neoplasm (e.g., lymphoma). leukemia (ALL), acute myeloid leukemia, acute lymphocytic In some embodiments the lymphoid neoplasm (e.g., lym 65 leukemia, and acute myelocytic leukemia (e.g., myeloblastic, phoma) is a B-cell neoplasm. Examples of B-cell neoplasms promyelocytic, myelomonocytic, monocytic, and erythroleu include, but are not limited to, precursor B-cell neoplasms kemia). US 8,911,786 B2 17 18 In some embodiments, there are provided methods of treat Soft tissue sarcoma, uterine Sacronomasynovioma, mesothe ing cancer with compositions comprising nanoparticles com lioma, Ewing's tumor, leiomyosarcoma, rhabdomyosar prising rapamycin or a derivative thereofanda carrier protein coma, coloncarcinoma, pancreatic cancer, breast cancer, ova (e.g., albumin), wherein the cancer is a liquid tumor or plas rian cancer, prostate cancer, Squamous cell carcinoma, basal macytoma. In some embodiments, there are provided meth cell carcinoma, adenocarcinoma, Sweat gland carcinoma, ods of treating cancer in an individual by administering to the sebaceous gland carcinoma, papillary carcinoma, papillary individual (e.g., a human) an effective amount of a composi adenocarcinomas, cystadenocarcinoma, medullary carci tion comprising nanoparticles comprising a rapamycinandan noma, bronchogenic carcinoma, renal cell carcinoma, albumin, wherein the cancer is a liquid tumor or plasmacy hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, toma. Plasmacytoma includes, but is not limited to, myeloma. 10 embryonal carcinoma, Wilm's tumor, cervical cancer, tes Myeloma includes, but is not limited to, an extramedullary ticular tumor, lung carcinoma, Small cell lung carcinoma, plasmacytoma, a solitary myeloma, and multiple myeloma. bladder carcinoma, epithelial carcinoma, glioma, astrocy In some embodiments, the plasmacytoma is multiple toma, medulloblastoma, craniopharyngioma, ependymoma, myeloma. pinealoma, hemangioblastoma, acoustic neuroma, oligoden In some embodiments, there are provided methods of treat 15 droglioma, menangioma, melanoma, neuroblastoma, and ret ing cancer in an individual by administering to the individual inoblastoma. (e.g., a human) an effective amount of a composition com Accordingly, in some embodiments, there are provided prising nanoparticles comprising rapamycin or a derivative methods of treating cancer in an individual by administering thereof and a carrier protein (e.g., albumin), wherein the to the individual (e.g., a human) an effective amount of a cancer is multiple myeloma. In some embodiments, there are composition comprising nanoparticles comprising rapamy provided methods of treating cancer in an individual by cin or a derivative thereofanda carrier protein (e.g., albumin), administering to the individual (e.g., a human) an effective wherein the cancer is breast cancer. In some embodiments, amount of a composition comprising nanoparticles compris there are provided a method of treating breast cancer in an ing a rapamycin and an albumin, wherein the cancer is mul individual by administering to the individual (e.g., a human) tiple myeloma. Examples of multiple myeloma include, but 25 an effective amount of a composition comprising nanopar are not limited to, IgG multiple myeloma, IgA multiple ticles that comprise rapamycin or a derivative thereof and an myeloma, Ig|D multiple myeloma, IgE multiple myeloma, albumin. In some embodiments, the breast cancer is early and nonsecretory multiple myeloma. In some embodiments, stage breast cancer, non-metastatic breast cancer, advanced the multiple myeloma is IgG multiple myeloma. In some breast cancer, stage IV breast cancer, locally advanced breast embodiments, the multiple myeloma is IgA multiple 30 cancer, metastatic breast cancer, breast cancer in remission, myeloma. In some embodiments, the multiple myeloma is a breast cancer in an adjuvant setting, or breast cancer in a smoldering or indolent multiple myeloma. In some embodi neoadjuvant setting. In some specific embodiments, the ments, the multiple myeloma is progressive multiple breast cancer is in a neoadjuvant setting. In some embodi myeloma. In some embodiments, multiple myeloma may be ments, there are provided methods of treating cancer at resistant to a drug, such as, but not limited to, , 35 advanced stage(s). In some embodiments, there are provided dexamethasone (Dex-), doxorubicin (DOX-), and methods of treating breast cancer (which may be HER2 posi (LR). tive or HER2 negative), including, for example, advanced In some embodiments, the individual may be a human who breast cancer, stage IV breast cancer, locally advanced breast has a gene, genetic mutation, or polymorphism associated cancer, and metastatic breast cancer. In some embodiments, with multiple myeloma (e.g., ras, PTEN, Rb1, MTS1/ 40 the individual may be a human who has a gene, genetic p16INK4A/ CDKN2, MTS2/p15INK4B, and/or p53) or has mutation, or polymorphism associated with breast cancer one or more extra copies of a gene associated with multiple (e.g., BRCA1, BRCA2, ATM, CHEK2, RAD51, AR, myeloma. In some embodiments, the individual has a ras or DIRAS3, ERBB2, TP53, AKT, PTEN, and/or PI3K) or has PTEN mutation. In some embodiments, the cancer cells are one or more extra copies of a gene (e.g., one or more extra dependent on an mTOR pathway to translate one or more 45 copies of the HER2 gene) associated with breast cancer. In mRNAs. In some embodiments, the cancer cells are not some embodiments, the method further comprises identify capable of synthesizing mRNAs by an mTOR-independent ing a cancer patient population (i.e. breast cancer population) pathway. In some embodiments, the cancer cells have based on a hormone receptor status of patients having tumor decreased or no PTEN activity or have decreased or no tissue not expressing both ER and PgR and administering to expression of PTEN compared to non-cancerous cells. In 50 the patient population an effective amount of a composition Some embodiments, the cancer cells have increased AKT comprising nanoparticles comprising rapamycin or a deriva activity and/or expression compared to non-cancerous cells. tive thereof and a carrier protein (e.g., albumin) In some embodiments, there are provided methods of treat In some embodiments, there are provided methods of treat ing cancer in an individual by administering to the individual ing cancer in an individual by administering to the individual (e.g., a human) an effective amount of a composition com 55 (e.g., a human) an effective amount of a composition com prising nanoparticles of rapamycin or a derivative thereofand prising nanoparticles comprising rapamycin or a derivative a carrier protein (e.g., albumin), wherein the cancer is a Solid thereof and a carrier protein (e.g., albumin), wherein the tumor. In some embodiments, there are provided methods of cancer is a renal cell carcinoma (also called kidney cancer, treating cancer in an individual by administering to the indi renal adenocarcinoma, or hypernephroma). In some embodi vidual (e.g., a human) an effective amount of a composition 60 ments, there are provided methods of treating cancer by comprising nanoparticles comprising a rapamycin and an administering to the individual (e.g., a human) an effective albumin, wherein the cancer is a solid tumor. In some embodi amount of a composition comprising nanoparticles compris ments, the Solid tumor includes, but is not limited to, sarco ing an albumin, wherein the cancer is a renal cell carcinoma. mas and carcinomas Such as fibrosarcoma, myxosarcoma, In some embodiments, the renal cell carcinoma is an adeno liposarcoma, chondrosarcoma, osteogenic sarcoma, chor 65 carcinoma. In some embodiments, the renal cell carcinoma is doma, angiosarcoma, endotheliosarcoma, lymphangiosar a clear cell renal cell carcinoma, papillary renal cell carci coma, lymphangioendotheliosarcoma, Kaposi's sarcoma, noma (also called chromophilic renal cell carcinoma), chro US 8,911,786 B2 19 20 mophobe renal cell carcinoma, collecting duct renal cell car limited to, large-cell carcinoma (e.g., large-cell neuroendo cinoma, granular renal cell carcinoma, mixed granular renal crine carcinoma, combined large-cell neuroendocrine carci cell carcinoma, renal angiomyolipomas, or spindle renal cell noma, basaloid carcinoma, lymphoepithelioma-like carci carcinoma. In some embodiments, the individual may be a noma, clear cell carcinoma, and large-cell carcinoma with human who has a gene, genetic mutation, or polymorphism rhabdoid phenotype), adenocarcinoma (e.g., acinar, papillary associated with renal cell carcinoma (e.g., VHL, TSC1. (e.g., bronchioloalveolar carcinoma, nonmucinous, muci TSC2, CUL2, MSH2, MLH1, INK4a/ARF, MET, TGF-C., nous, mixed mucinous and nonmucinous and indeterminate TGF-B1, IGF-1, IGF-1R, AKT, and/or PTEN) or has one or cell type), Solid adenocarcinoma with mucin, adenocarci more extra copies of a gene associated with renal cell carci noma with mixed subtypes, well-differentiated fetal adeno noma. In some embodiments, the renal cell carcinoma is 10 associated with (1) von Hippel-Lindau (VHL) syndrome, (2) carcinoma, mucinous (colloid) adenocarcinoma, mucinous hereditary papillary renal carcinoma (HPRC), (3) familial cystadenocarcinoma, signet ring adenocarcinoma, and clear renal oncocytoma (FRO) associated with Birt–Hogg–Dube cell adenocarcinoma), neuroendocrine lung tumors, and syndrome (BHDS), or (4) hereditary renal carcinoma (HRC). squamous cell carcinoma (e.g., papillary, clear cell, Small There are provided methods of treating renal cell carcinoma 15 cell, and basaloid). In some embodiments, the NSCLC may at any of the four stages, I, II, III, or IV, according to the be, according to TNM classifications, a stage T tumor (pri American Joint Committee on Cancer (AJCC) staging mary tumor), a stage N tumor (regional lymph nodes), or a groups. In some embodiments, the renal cell carcinoma is stage M. tumor (distant metastasis). In some embodiments, stage IV renal cell carcinoma. the lung cancer is a carcinoid (typical or atypical), adenos In some embodiments, there are provided methods of treat quamous carcinoma, cylindroma, or carcinoma of the Sali ing cancer in an individual by administering to the individual vary gland (e.g., adenoid cystic carcinoma or mucoepider (e.g., a human) an effective amount of a composition com moid carcinoma). In some embodiments, the lung cancer is a prising nanoparticles comprising rapamycin or a derivative carcinoma with pleomorphic, sarcomatoid, or sarcomatous thereof and a carrier protein (e.g., albumin), wherein the elements (e.g., carcinomas with spindle and/or giant cells, cancer is prostate cancer. In some embodiments, there are 25 spindle cell carcinoma, giant cell carcinoma, carcinosar provided methods of treating cancer by administering to the coma, or pulmonary blastoma). In some embodiments, the individual (e.g., a human) an effective amount of a composi cancer is Small cell lung cancer (SCLC; also called oat cell tion comprising nanoparticles comprising a rapamycinandan carcinoma). The Small cell lung cancer may be limited-stage, albumin, wherein the cancer is prostate cancer. In some extensive stage or reoccurent Small cell lung cancer. In some embodiments, the prostate cancer is an adenocarcinoma. In 30 embodiments, the individual may be a human who has a gene, Some embodiments, the prostate cancer is a sarcoma, neu genetic mutation, or polymorphism suspected or shown to be roendocrine tumor, small cell cancer, ductal cancer, or a lym associated with lung cancer (e.g., SASH1, LATS1, IGF2R, phoma. There are provided methods of treating prostate can PARK2, KRAS, PTEN, Kras2, Krag, Pas1, ERCC1, XPD, cer at any of the four stages, A, B, C, or D, according to the IL8RA, EGFR, C-AD, EPHX, MMP1, MMP2, MMP3, Jewett staging system. In some embodiments, the prostate 35 MMP12, IL1B, RAS, and/or AKT) or has one or more extra cancer is stage A prostate cancer (The cancer cannot be felt copies of a gene associated with lung cancer. during a rectal exam.). In some embodiments, the prostate In some embodiments, there are provided methods of treat cancer is stage B prostate cancer (The tumor involves more ing cancer in an individual by administering to the individual tissue within the prostate, it can be felt during a rectal exam, (e.g., a human) an effective amount of a composition com or it is found with a biopsy that is done because of a high PSA 40 prising nanoparticles comprising rapamycin or a derivative level.). In some embodiments, the prostate cancer is stage C thereof and a carrier protein (e.g., albumin), wherein the prostate cancer (The cancer has spread outside the prostate to cancer is brain cancer. In some embodiments, there are pro nearby tissues.). In some embodiments, the prostate cancer is vided methods of treating cancer by administering to the stage D prostate cancer. In some embodiments, the prostate individual (e.g., a human) an effective amount of a composi cancer may be androgen independent prostate cancer (AIPC). 45 tion comprising nanoparticles comprising a rapamycin and an In some embodiments, the prostate cancer may be androgen albumin, wherein the cancer is brain cancer. In some embodi dependent prostate cancer. In some embodiments, the pros ments, the brain cancer is glioma, brain stem glioma, cerebel tate cancer may be refractory to hormone therapy. In some lar or cerebral astrocytoma (e.g., pilocytic astrocytoma, dif embodiments, the prostate cancer may be substantially fuse astrocytoma, or anaplastic (malignant) astrocytoma), refractory to hormone therapy. In some embodiments, the 50 malignant glioma, ependymoma, oligodenglioma, menin individual may be a human who has a gene, genetic mutation, gioma, craniopharyngioma, haemangioblastomas, medullo or polymorphism associated with prostate cancer (e.g., RNA blastoma, Supratentorial primitive neuroectodermal tumors, SEL/HPC1, ELAC2/HPC2, SR-A/MSR1, CHEK2, BRCA2, visual pathway and hypothalamic glioma, or glioblastoma. In PON1, OGG1, MICA-1, TLR4, and/or PTEN) or has one or Some embodiments, the brain cancer is glioblastoma (also more extra copies of a gene associated with prostate cancer. 55 called glioblastoma multiforme or grade 4 astrocytoma). In In some embodiments, there are provided methods of treat Some embodiments, the glioblastoma is radiation-resistant. ing cancer in an individual by administering to the individual In some embodiments, the glioblastoma is radiation-sensi (e.g., a human) an effective amount of a composition com tive. In some embodiments, the glioblastoma may be infrat prising nanoparticles comprising rapamycin or a derivative entorial. In some embodiments, the glioblastoma is Supraten thereof and a carrier protein (e.g., albumin), wherein the 60 torial. In some embodiments, the individual may be a human cancer is lung cancer. In some embodiments, there are pro who has a gene, genetic mutation, or polymorphism associ vided methods of treating cancer by administering to the ated with brain cancer (e.g., glioblastoma) (e.g., NRPB, individual (e.g., a human) an effective amount of a composi MAGE-E1, MMACI-E1, PTEN, LOH, p53, MDM2, DCC, tion comprising nanoparticles comprising a rapamycinandan TP-73, Rb1, EGFR, PDGFR-O, PMS2, MLH1, and/or albumin, wherein the cancer is lung cancer. In some embodi 65 DMBT1) or has one or more extra copies of a gene associated ments, the cancer is lung cancer is a non-Small cell lung with brain cancer (e.g., glioblastoma) (e.g., MDM2, EGFR, cancer (NSCLC). Examples of NCSLC include, but are not and PDGR-C). US 8,911,786 B2 21 22 In some embodiments, there are provided methods of treat thereof and a carrier protein (e.g., albumin), wherein the ing cancer in an individual by administering to the individual cancer is a neuroendocrine cancer. In some embodiments, the (e.g., a human) an effective amount of a composition com individual may be a human who has a gene, genetic mutation, prising nanoparticles comprising rapamycin or a derivative or polymorphism associated with neuroendocrine cancer thereof and a carrier protein (e.g., albumin), wherein the (e.g., TSC1, TSC2, IGF-1, IGF-1R, and/or VHL) or has one cancer is melanoma. In some embodiments, there are pro or more extra copies of a gene associated with neurondocrine vided methods of treating cancer by administering to the CaCC. individual (e.g., a human) an effective amount of a composi In some embodiments, there are provided methods of treat tion comprising nanoparticles comprising a rapamycinandan ing cancer in an individual by administering to the individual albumin, wherein the cancer is melanoma. 10 (e.g., a human) an effective amount of a composition com In some embodiments, there are provided methods of treat prising nanoparticles comprising rapamycin or a derivative ing cancer in an individual by administering to the individual thereof and a carrier protein (e.g., albumin), wherein the (e.g., a human) an effective amount of a composition com cancer is colon cancer. In some embodiments, the individual prising nanoparticles comprising rapamycin or a derivative may be a human who has a gene, genetic mutation, or poly thereof and a carrier protein (e.g., albumin), wherein the 15 morphism associated with colon cancer (e.g., RAS, AKT, cancer is ovarian cancer. In some embodiments, there are PTEN, PI3K, and/or EGFR) or has one or more extra copies provided methods of treating cancer by administering to the of a gene associated with colon cancer. individual (e.g., a human) an effective amount of a composi In some embodiments, there are provided methods of treat tion comprising nanoparticles comprising a rapamycinandan ing cancer in an individual by administering to the individual albumin, wherein the cancer is ovarian cancer. In some (e.g., a human) an effective amount of a composition com embodiments, the cancer is ovarian epithelial cancer. Exem prising nanoparticles comprising rapamycin or a derivative plary ovarian epithelial cancer histological classifications thereof and a carrier protein (e.g., albumin), wherein the include: Serous cystomas (e.g., serous benign cystadenomas, cancer is characterized by PI3K and/or AKT activation. In serous cystadenomas with proliferating activity of the epithe some embodiments, the cancer characterized by PI3K and/or lial cells and nuclear abnormalities but with no infiltrative 25 AKT activation is HER2" breast cancer, chronic myelog destructive growth, or serous cystadenocarcinomas), muci enous leukemia, ovarian cancer, endometrial cancer, sar nous cystomas (e.g., mucinous benign cystadenomas, muci coma, squamous cell carcinoma of the head and neck, or nous cystadenomas with proliferating activity of the epithe thyroid cancer. In some variations, the cancer is further char lial cells and nuclear abnormalities but with no infiltrative acterized by AKT gene amplification. destructive growth, or mucinous cystadenocarcinomas), 30 In some embodiments, there are provided methods of treat endometrioid tumors (e.g., endometrioid benign cysts, ing cancer in an individual by administering to the individual endometrioid tumors with proliferating activity of the epithe (e.g., a human) an effective amount of a composition com lial cells and nuclear abnormalities but with no infiltrative prising nanoparticles comprising rapamycin or a derivative destructive growth, or endometrioid adenocarcinomas), clear thereof and a carrier protein (e.g., albumin), wherein the cell (mesonephroid) tumors (e.g., begin clear cell tumors, 35 cancer is characterized by cyclin D1 overexpression. In some clear cell tumors with proliferating activity of the epithelial embodiments, the cancer characterized by cyclin Doverex cells and nuclear abnormalities but with no infiltrative pression is mantle cell lymphoma or breast cancer. destructive growth, or clear cell cystadenocarcinomas), In some embodiments, there are provided methods of treat unclassified tumors that cannot be allotted to one of the above ing cancer in an individual by administering to the individual groups, or other malignant tumors. In various embodiments, 40 (e.g., a human) an effective amount of a composition com the ovarian epithelial cancer is stage I (e.g., stage IA, IB, or prising nanoparticles comprising rapamycin or a derivative IC), stage II (e.g., stage IIA, IIB, or IIC), stage III (e.g., stage thereof and a carrier protein (e.g., albumin), wherein the IIIA, IIIB, or IIIC), or stage IV. In some embodiments, the cancer is characterized by cMYC overexpression. In some individual may be a human who has a gene, genetic mutation, embodiments, the cancer characterized by cMYC overex or polymorphism associated with ovarian cancer (e.g., 45 pression is Burkitt lymphoma. BRCA1 or BRCA2) or has one or more extra copies of a gene In some embodiments, there are provided methods of treat associated with ovarian cancer (e.g., one or more extra copies ing cancer in an individual by administering to the individual of the HER2 gene). (e.g., a human) an effective amount of a composition com In some embodiments, the cancer is an ovarian germ cell prising nanoparticles comprising rapamycin or a derivative tumor. Exemplary histologic Subtypes include dysgermino 50 thereof and a carrier protein (e.g., albumin), wherein the mas or other germ cell tumors (e.g., endodermal sinus tumors cancer is characterized by HIF overexpression. In some Such as hepatoid or intestinal tumors, embryonal carcinomas, embodiments, the cancer characterized by HIF overexpres olyembryomas, choriocarcinomas, teratomas, or mixed form sion is renal cell carcinoma or Von Hippel-Lindau. In some tumors). Exemplary teratomas are immature teratomas, embodiments, the cancer further comprises a VHL. mutation. mature teratomas, Solid teratomas, and cystic teratomas (e.g., 55 In some embodiments, there are provided methods of treat dermoid cysts Such as mature cystic teratomas, and dermoid ing cancer in an individual by administering to the individual cysts with malignant transformation). Some teratomas are (e.g., a human) an effective amount of a composition com monodermal and highly specialized, such as struma ovarii, prising nanoparticles comprising rapamycin or a derivative carcinoid, struma ovarii and carcinoid, or others (e.g., malig thereof and a carrier protein (e.g., albumin), wherein the nant neuroectodermal and ependymomas). In some embodi 60 cancer is characterized by TSC1 and/or TSC2 loss. In some ments, the ovarian germ cell tumor is stage I (e.g., stage IA, embodiments, the cancer characterized by TSC1 and/or IB, or IC), stage II (e.g., stage IIA, IIB, or IIC), stage III (e.g., TSC2 is tuberous Sclerosis or pulmonary lymphangiomyo stage IIIA, IIIB, or IIIC), or stage IV. matosis. In some embodiments, there are provided methods of treat In some embodiments, there are provided methods of treat ing cancer in an individual by administering to the individual 65 ing cancer in an individual by administering to the individual (e.g., a human) an effective amount of a composition com (e.g., a human) an effective amount of a composition com prising nanoparticles comprising rapamycin or a derivative prising nanoparticles comprising rapamycin or a derivative US 8,911,786 B2 23 24 thereof and a carrier protein (e.g., albumin), wherein the are not limited to, age, sex, race, diet, history of previous cancer is characterized by a TSC2 mutation. In some embodi disease, presence of precursor disease, genetic (e.g., heredi ments, the cancer characterized by TSC2 mutation is renal tary) considerations, and environmental exposure. In some angiomyolipomas. embodiments, the individuals at risk for cancer include, e.g., In some embodiments, there are provided methods of treat those having relatives who have experienced this disease, and ing cancer in an individual by administering to the individual those whose risk is determined by analysis of genetic or (e.g., a human) an effective amount of a composition com biochemical markers. prising nanoparticles comprising rapamycin or a derivative Any of the methods of treatment provided herein may be thereof and a carrier protein (e.g., albumin), wherein the practiced in an adjuvant setting. Any of the methods of treat cancer is characterized by a PTEN mutation. In some embodi 10 ment provided herein may be practiced in a neoadjuvant ments, the PTEN mutation is a loss of PTEN function. In setting, i.e., the method may be carried out before the pri some embodiments, the cancer characterized by a PTEN mary/definitive therapy. In some embodiments, any of the mutation is glioblastoma, endometrial cancer, prostate can methods of treatment provided herein may be used to treat an cer, sarcoma, or breast cancer. individual who has previously been treated. Any of the meth In some embodiments, the methods of treatment provided 15 ods of treatment provided herein may be used to treat an herein may also be used to treat a cancer which is not a solid individual who has not previously been treated. Any of the tumor (i.e., other than a solid tumor). In some embodiments, methods of treatment provided herein may be used to treat an the methods of treatment provided herein may also be used to individual at risk for developing cancer, but has not been treat a cancer which is not a carcinoma. In some embodi diagnosed with cancer. Any of the methods of treatment pro ments, the methods of treatment provided herein may also be vided herein may be used as a first line therapy. Any of the used to treata cancer which is not a sarcoma. In some embodi methods of treatment provided herein may be used as a sec ments, the methods of treatment provided herein may also be ond line therapy. used to treat a cancer which is not a lymphoma. In some In some embodiments of any of the methods of treatment embodiments, the methods of treatment provided herein may provided herein, a taxane is not administered to the indi also be used to treat a cancer which is not colon cancer (i.e., 25 vidual. In some embodiments, the taxane administered is not other than colon cancer). In some embodiments, the methods a nanoparticle composition. In some embodiments, the nano of treatment provided herein may also be used to treat a particle composition comprising rapamycin or a derivative cancer which is not breast cancer (i.e., other than breast thereof is not administered in conjunction with a taxane. In cancer). In some embodiments, the methods of treatment Some embodiments, a taxane is not administered to the indi provided herein may also be used to treat a cancer which is not 30 vidual during the time period in which the individual is an ovarian cancer, brain cancer, and/or prostate cancer (i.e., receiving one or more doses of a nanoparticle composition other than ovarian cancer, brain cancer, and/or prostate can comprising rapamycin or a derivative thereof In some cer). embodiments, the individual was treated with a taxane before Any of the methods of treatment provided herein may be treatment begins with a nanoparticle composition comprising used to treat a primary tumor. Any of the methods of treatment 35 rapamycin or a derivative thereof For example, the individual provided herein may also be used to treat a metastatic cancer may have received a taxane one or more days, weeks, months, (that is, cancer that has metastasized from the primary tumor). or years before treatment begins with a nanoparticle compo Any of the methods of treatment provided herein may be used sition comprising rapamycin or a derivative thereof In other to treat cancer at an advanced stage. Any of the methods of embodiments, the individual never receives a taxane before treatment provided herein may be used to treat cancer at 40 treatment begins with a nanoparticle composition comprising locally advanced stage. Any of the methods of treatment rapamycin or a derivative thereof In some embodiments, the provided herein may be used to treat early stage cancer. Any individual is treated with a taxane after treatment with a of the methods of treatment provided herein may be used to nanoparticle composition comprising rapamycin or deriva treat cancer in remission. In some of the embodiments of any tive thereof terminates. In other embodiments, the individual of the methods of treatment provided herein, the cancer has 45 is never treated with a taxane after treatment with a nanopar reoccurred after remission. In some embodiments of any of ticle composition comprising rapamycin orderivative thereof the methods of treatment provided herein, the cancer is pro terminates. In some embodiments, the composition, first gressive cancer. Any of the methods of treatment provided therapy, and/or second therapy do not contain a taxane. In herein may be used to treat cancer substantially refractory to other embodiments, the composition, first therapy, and/or hormone therapy. Any of the methods of treatment provided 50 second therapy comprise a taxane. In some embodiments, the herein may be used to treat HER-2 positive cancer. Any of the first and/or second therapies do not comprise a SPARC methods of treatment provided herein may be used to treat polypeptide oranti-SPARC antibody (i.e., other than. SPARC HER-2 negative cancer. polypeptide or anti-SPARC antibody). Any of the methods of treatment provided herein may be Any of the methods of treatment provided herein may be used to treat and individual (e.g., human) who has been diag 55 used to treat, stabilize, prevent, and/or delay any type or stage nosed with or is suspected of having cancer. In some embodi of cancer. In some embodiments, the individual is at least ments, the individual may be a human who exhibits one or about any of 40, 45, 50,55, 60, 65,70, 75,80, or 85 years old. more symptoms associated with cancer. In some embodi In some embodiments, one or more symptoms of the cancer ments, the individual may have advanced disease or a lesser are ameliorated or eliminated. In some embodiments, the size extent of disease. Such as low tumor burden. In some embodi 60 of a tumor, the number of cancer cells, or the growth rate of a ments, the individual is at an early stage of a cancer. In some tumor decreases by at least about any of 10%. 20%, 30%, embodiments, the individual is at an advanced stage of can 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100%. In some cer. In some of the embodiments of any of the methods of embodiments, the cancer is delayed or prevented. treatment provided herein, the individual may be a human Combination Therapy who is genetically or otherwise predisposed (e.g., risk factor) 65 The present invention also features methods for the treat to developing cancer who has or has not been diagnosed with ment of cancer using combination therapies. Accordingly, in cancer. In some embodiments, these risk factors include, but Some embodiments, a second therapy useful for treating can US 8,911,786 B2 25 26 cer is also administered to the individual. In some embodi amount of a composition comprising nanoparticles compris ments, the second therapy includes Surgery, radiation, gene ing a rapamycin or a derivative thereof and a carrier protein therapy, immunotherapy, bone marrow transplantation, stem (e.g., albumin); and b) an effective amount of a composition cell transplantation, hormonetherapy, targeted therapy, cryo comprising nanoparticles comprising at least one other che therapy, ultrasound therapy, photodynamic therapy, and/or motherapeutic agent and a carrier protein (such as albumin). chemotherapy (e.g., one or more compounds or pharmaceu In some embodiments, the method comprises administering tically acceptable salts thereofuseful for treating cancer). It is to the individual a) an effective amount of a composition understood that reference to and description of methods of comprising nanoparticles comprising a rapamycin and an treating cancer above is exemplary and that the description albumin; and b) an effective amount of a composition com applies equally to and includes methods of treating cancer 10 prising nanoparticles comprising at least one other chemo using combination therapy. therapeutic agent and a carrier protein (Such as albumin). In In one Such aspect, the invention provides a method of Some embodiments, the chemotherapeutic agent is any of treating cancer in an individual by administering to the indi (and in some embodiments selected from the group consist vidual an effective amount of a combination of a) a first ing of) thiocolchicine or its derivatives (such as dimeric thio therapy that includes a composition comprising nanoparticles 15 colchicine, including for example nab-5404, nab-5800, and that include rapamycin or a derivative thereof and a carrier nab-5801), and geldanamycin or its derivatives (such as protein (e.g., albumin) and b) a second therapy useful for 17-allyl amino geldanamycin (17-AAG)). In some embodi treating cancer. In some embodiments, the second therapy ments, the chemotherapeutic agent is a taxane or a derivative includes Surgery, radiation, gene therapy, immunotherapy, thereof (e.g., paclitaxel, , and ). In some bone marrow transplantation, stem cell transplantation, hor embodiments, the chemotherapeutic agent is not a taxane. In mone therapy, targeted therapy, cryotherapy, ultrasound other embodiments, the chemotherapeutic is not a taxane. In therapy, photodynamic therapy, and/or chemotherapy (e.g., Some embodiments, the chemotherapeutic agent is 17-AAG. one or more compounds useful for treating cancer). In some In Some embodiments, the chemotherapeutic agent is dimeric embodiments, the first and/or second therapies do not include thiocolchicine. a taxane. In other embodiments, the first and/or second thera 25 An exemplary and non-limiting list of chemotherapeutic pies do include a taxane. In some embodiments, the first agents contemplated is provided herein. Suitable chemo and/or second therapies do not comprise a SPARC polypep therapeutic agents include, for example, Vinca alkaloids, tide or anti-SPARC antibody. agents that disrupt formation (such as colchi In some embodiments, the invention provides methods of cines and its derivatives), anti-angiogenic agents, therapeutic treating cancer in an individual, comprising administering to 30 antibodies, EGFR targeting agents, tyrosine kinase targeting the individuala) an effective amount of a composition com agent (Such as tyrosine kinase inhibitors), transitional metal prising nanoparticles comprising a rapamycin or a derivative complexes, proteasome inhibitors, antimetabolites (such as thereof and a carrier protein (e.g., albumin); and b) an effec nucleoside analogs), alkylating agents, platinum-based tive amount of at least one other chemotherapeutic agent. In agents, anthracycline antibiotics, topoisomerase inhibitors, Some embodiments, the nanoparticles comprise rapamycin 35 therapeutic antibodies, (such as all-trans retinoic and an albumin. In some embodiments, the chemotherapeutic acids or a derivatives thereof); geldanamycin or a derivative agent is any of (and in Some embodiments selected from the thereof (such as 17-AAG), and other standard chemothera group consisting of) agents (including nucleo peutic agents well recognized in the art. side analogs), platinum-based agents, alkylating agents, In some embodiments, the chemotherapeutic agent is any tyrosine kinase inhibitors, anthracycline antibiotics, Vinca 40 of (and in some embodiments selected from the group con alkloids, proteasome inhibitors, , modulators of sisting of) adriamycin, colchicine, , acti HER2/neu (such as inhibitors of HER2/neu for example Her nomycin, , duanorubicin, doxorubicin, , ceptinR), modulators of EGFR (such as inhibitors of EGFR mitomycin, methotrexate, , fluorouracil, carbo for example ErbituX(R), modulators of VEGFR, farnosyl platin, (BCNU), methyl CCNU, cisplatin, etopo transferase inhibitors, and topoisomerase inhibitors. In some 45 side, epotetin alfa, interferons (e.g., IFN-C), embodiments, the chemotherapeutic is not a taxane (i.e., the and derivatives thereof, letrozole, panitumumab (Vectibix(R), compound is a chemotherapeutic agent other than a taxane). phenesterine, topetecan, , , tamoxifen, Preferred drug combinations for sequential or co-administra thalidomide, (Zarnestra(R), piposulfan, nab-5404, tion or simultaneous administration with nanoparticles com nab-5800, nab-5801, , HKP Ortataxel, gemcitab prising a rapamycin or a derivative thereof and a carrier pro 50 ine, Herceptin R, , Doxil R, capecitabine, tein (e.g., albumin) are those which show enhanced AlimtaR), Avastin R, Velcade(R), Tarceva(R), NeulastaR), Lapa anticancer activity when compared with the single compo tinib, Sorafenib, derivatives thereof, chemotherapeutic nents alone, especially combinations that lead to regression of agents known in the art, and the like. In some embodiments, cancer and/or cure from cancer. the chemotherapeutic agent is a composition comprising The chemotherapeutic agents described herein can be the 55 nanoparticles comprising a thiocolchicine derivative and a agents themselves, pharmaceutically acceptable salts thereof, carrier protein (such as albumin). In some embodiments, the and pharmaceutically acceptable esters thereof, as well as chemotherapeutic agent is a taxane or a derivative thereof Stereoisomers, enantiomers, racemic mixtures, and the like. (e.g., paclitaxel, docetaxel, and ortataxel). In some embodi The chemotherapeutic agent or agents as described can be ments, the chemotherapeutic is not a taxane. administered as well as a pharmaceutical composition con 60 In some embodiments, the chemotherapeutic agent is a taining the agent(s), wherein the pharmaceutical composition antineoplastic agent including, but is not limited to, carbopl comprises a pharmaceutically acceptable carrier vehicle, or atin, NavelbineR (vinorelbine), anthracycline (Doxil R), the like. lapatinib (GW57016), Herceptin R, gemcitabine (Gemzar(R), The chemotherapeutic agent may be present in a nanopar capecitabine (Xeloda(R), Alimta R, cisplatin, 5-fluorouracil, ticle composition. For example, in Some embodiments, there 65 epirubicin, cyclophosphamide, Avastin R, VelcadeR), etc. is provided a method of treating cancer in an individual, In some embodiments, the chemotherapeutic agent is an comprising administering to the individual a) an effective antagonist of other factors that are involved in tumor growth, US 8,911,786 B2 27 28 such as EGFR, ErbB2 (also known as Herb), ErbB3, ErbB4, the tyrosine kinase inhibitor is an inhibitor of BCR-Abl. In or TNF. Sometimes, it may be beneficial to also administer Some embodiments, the tyrosine kinase inhibitor is an inhibi one or more cytokines to the individual. In some embodi tor of IGF-1R. ments, the therapeutic agent is a growth inhibitory agent. In Some embodiments, the method is for treating non-small Suitable dosages for the growth inhibitory agent are those cell lung carcinoma. In some embodiments, the method is for presently used and may be lowered due to the combined treating brain cancer (e.g., glioblastoma). In some embodi action (synergy) of the growth inhibitory agent and the rapa ments, the method is for treating colorectal cancer, gas mycin or derivative thereof. In some embodiments, the che trointestinal stromal tumor, prostate cancer, ovarian cancer, motherapeutic agent is a chemotherapeutic agent other than or thyroid cancer. In some embodiments, the method is for 10 treatment of prostate cancer (e.g., advanced prostate cancer). an anti-VEGF antibody, a HER2 antibody, interferon, and an In some embodiments, the method is for treatment of breast HGFB antagonist. cancer, including treatment of metastatic breast cancer and Reference to a chemotherapeutic agent herein applies to treatment of breast cancer in a neoadjuvant setting. In some the chemotherapeutic agent or its derivatives and accordingly embodiments, the method is for treatment of advanced solid the invention contemplates and includes either of these 15 tumor. In some embodiments, the method is for treatment of embodiments (agent; agent orderivative(s)). “Derivatives” of multiple myeloma. In some embodiments, the method com a chemotherapeutic agent or other chemical moiety include, prises simultaneous and/or sequential administration of at but are not limited to, compounds that are structurally similar least one EGFR blocker, inhibitor, or antagonist. In some to the chemotherapeutic agent or moiety, compounds that are embodiments, the individual has activating mutation(s) in the in the same general chemical class as the chemotherapeutic kinase domain of EGFR. In some embodiments, the indi agent or moiety, analogs of chemotherapeutic agents, or phar vidual is of Asian or East Asian ancestry. In some embodi maceutically acceptable salts of chemotherapeutic agents or ments, the individual is female. their derivatives. In some embodiments, the derivative of the In some embodiments, the invention provides a method of chemotherapeutic agent or moiety retains similar chemical treating cancer in an individual, comprising administering to and/or physical property (including, for example, functional 25 the individuala) an effective amount of a composition com ity) of the chemotherapeutic agent or moiety. prising nanoparticles comprising a rapamycin or a derivative In some embodiments, the invention provides a method of thereof and a carrier protein (e.g., albumin), and b) an effec treating cancer in an individual, comprising administering to tive amount of an antimetabolite agent (Such as a nucleoside the individuala) an effective amount of a composition com analog, including for example purine analogs and pyrimidine prising nanoparticles comprising a rapamycin or a derivative 30 analogs). In some embodiments, the invention provides a thereof and a carrier protein (e.g., albumin), and b) an effec method of treating cancer in an individual, comprising tive amount of a tyrosine kinase inhibitor. In some embodi administering to the individual a) an effective amount of a ments, the invention provides a method of treating cancer in composition comprising nanoparticles comprising rapamy an individual, comprising administering to the individual a) cin and an albumin, and b) an effective amount of an antime an effective amount of a composition comprising nanopar 35 tabolite agent. An 'antimetabolic agent' is an agent which is ticles comprising rapamycin and an albumin, and b) an effec structurally similar to a metabolite, but cannot be used by the tive amount of a tyrosine kinase inhibitor. Suitable tyrosine body in a productive manner. Many antimetabolite agents kinase inhibitors include, for example, imatinib (GleevecR), interfere with production of nucleic acids, RNA and DNA. nilotinim, gefitinib (Iressa R, ZD-1839), erlotinib (Tarceva(R): For example, the antimetabolite can be a nucleoside analog, OSI-774), Sunitinib malate (Sutent(R), Sorafenib (Nexavar(R), 40 which includes, but is not limited to, , azathio and Lapatinib (GW562016; Tykerb). In some embodiments, prine, capecitabine (XelodaR), , , the tyrosine kinase inhibitor is a multiple reversible ErbB1 cytosine arabinoside (ara-C, cytosar), , fluorou family tyrosine kinase inhibitor (e.g., laptinib). In some racil (such as 5-fluorouracil), 9-(2-phosphonylmethoxyethyl) embodiments, the tyrosine kinase inhibitor is a single revers adenine, UFT hydoxyurea, gemcitabine, , ible EGFR tyrosine kinase inhibitor (e.g., gefitinib or erlo 45 methotrexate, thioguanine (such as 6-thioguanine). Other tinib). In some embodiments, the tyrosine kinase inhibitor is anti-metabolites include, for example, L- (El erlotinib. In some embodiments, the tyrosine kinase inhibitor spa), decarbazine (DTIC), 2-deoxy-D-glucose, and procarba is gefitinib. In some embodiments, the tyrosine kinase inhibi Zine (matulane). In some embodiments, the nucleoside ana tor is a single irreversible EGFR tyrosine kinase inhibitor log is any of (and in some embodiments selected from the (e.g., EKB-569 or CL-387,785). In some embodiments, the 50 group consisting of) gemcitabine, fluorouracil, and capecit tyrosine kinase inhibitor is a multiple irreversible ErbB fam abine. In some embodiments, the method is for treatment of ily tyrosine kinase inhibitor (e.g. canertinib (CL-1033; metastatic breast cancer or locally advanced breast cancer. In PD183805), HKI-272, BIBW 2992, or HKI-357). In some some embodiments, the method is for first line treatment of embodiments, the tyrosine kinase inhibitor is a multiple metastatic breast cancer. In some embodiments, the method is reversible tyrosine kinase inhibitor (e.g., ZD-6474, ZD-6464, 55 for treatment of breast cancer in a neoadjuvant setting. In AEE 788, or XL647). In some embodiments, the tyrosine some embodiments, the method is for treatment of any of kinase inhibitor inhibits ErbB family heterodimerization NSCLC, metastatic colorectal cancer, pancreatic cancer, or (e.g., BMS-599626). In some embodiments, the tyrosine advanced Solid tumor. kinase inhibitor inhibits protein folding by affecting HSP90 In some embodiments, the invention provides a method of (e.g., benzoquinone ansamycin, IPI-504, or 17-AAG). In 60 treating cancer in an individual, comprising administering to some embodiments, there is provided a method to inhibit the the individuala) an effective amount of a composition com proliferation of EGFR expressing tumors in a mammal com prising nanoparticles comprising a rapamycin or a derivative prising administering to a mammal infected with Such tumors thereof and a carrier protein (e.g., albumin), and b) an effec an effective amount of a composition comprising nanopar tive amount of an alkylating agent. In some embodiments, the ticles comprising a rapamycin or a derivative thereof and a 65 invention provides a method of treating cancer in an indi carrier protein (e.g., albumin) and gefitinib, wherein the gefi vidual, comprising administering to the individuala) an effec tinib is administered by pulse-dosing. In some embodiments, tive amount of a composition comprising nanoparticles com US 8,911,786 B2 29 30 prising rapamycin and an albumin, and b) an effective amount tive amount of a Vinca alkloid. In some embodiments, the of an alkylating agent. Suitable alkylating agents include, but invention provides a method of treating cancer in an indi are not limited to, cyclophosphamide (Cytoxan), mechlore vidual, comprising administering to the individuala) an effec thamine, , melphalan, carmustine (BCNU), tive amount of a composition comprising nanoparticles com , , alkyl Sulphonates, ethylene imines, nitro prising rapamycin and an albumin, and b) an effective amount gen mustard analogs, estramustine Sodium phosphate, ifos of a Vinca alkloid. Suitable Vinca alkaloids include, for famide, , , and streptozocin. In some example, vinblastine, Vincristine, , vinorelbine (Na embodiments, the alkylating agent is cyclophosphamide. In Velbine(R), and VP-16. In some embodiments, the Vinca alka Some embodiments, the cyclophosphamide is administered loid is vinorelbine (Navelbine(R). In some embodiments, the prior to the administration of the nanoparticle composition. In 10 some embodiments, the method is for treatment of an early method is for treatment of stage IV breast cancer and lung stage breast cancer. In some embodiments, the method is for CaCC. treatment of a breast cancer in an adjuvant or a neoadjuvant In some embodiments, the invention provides a method of Setting. treating cancer in an individual, comprising administering to In some embodiments, the invention provides a method of 15 the individuala) an effective amount of a composition com treating cancer in an individual, comprising administering to prising nanoparticles comprising a rapamycin or a derivative the individuala) an effective amount of a composition com thereof and a carrier protein (e.g., albumin), and b) an effec prising nanoparticles comprising a rapamycin or a derivative tive amount of a . In some embodi thereof and a carrier protein (e.g., albumin), and b) an effec ments, the invention provides a method of treating cancer in tive amount of a platinum-based agent. In some embodi an individual, comprising administering to the individual a) ments, the invention provides a method of treating cancer in an effective amount of a composition comprising nanopar an individual, comprising administering to the individual a) ticles comprising rapamycin and an albumin, and b) an effec an effective amount of a composition comprising nanopar tive amount of a topoisomerase inhibitor. In some embodi ticles comprising rapamycin and an albumin, and b) an effec ments, the chemotherapeutic agent is a topoisomerase tive amount of a platinum-based agent. Suitable platinum 25 inhibitor, including, for example, inhibitor of topoisomerase based agents include, but are not limited to, carboplatin, I and topoisomerase II. Exemplary inhibitors of topoi cisplatin, and . In some embodiments, the plati Somerase I include, but are not limited to, camptothecin, Such num-based agent is carboplatin. In some embodiments, the as irinotecan and . Exemplary inhibitors of topoi platinum-based agent is oxaliplatin. We have observed that Somerase II include, but are not limited to, , etopo rapamycin inhibited oxaliplatin induced apoptosis in a dose 30 side, phosphate, and . dependent manner. This inhibition was not overwhelmed by increasing amount of oxaliplatin up to 1:1 (w/w) ratio of the In some embodiments, the invention provides a method of two drugs. The same was observed for Eloxatin R (oxaliplatin treating cancer in an individual, comprising administering to injection). the individuala) an effective amount of a composition com prising nanoparticles comprising a rapamycin or a derivative In some embodiments, the method is for treatment of breast 35 cancer (HER2 positive or HER2 negative, including meta thereof and a carrier protein (e.g., albumin), and b) an effec static breast cancer and advanced breast cancer); lung cancer tive amount of an antiangiogenic agent. In some embodi (including advanced NSCLC, first line NSCLC, SCLC, and ments, the invention provides a method of treating cancer in advanced solid tumor malignancies in the lung); ovarian can an individual, comprising administering to the individual a) cer; head and neck cancer, and melanoma (including meta 40 an effective amount of a composition comprising nanopar static melanoma). ticles comprising rapamycin and an albumin, and b) an effec In some embodiments, the invention provides a method of tive amount of an antiangiogenic agent. In some embodi treating cancer in an individual, comprising administering to ments, the method is for treatment of metastatic breast cancer, the individuala) an effective amount of a composition com breast cancer in an adjuvant setting or a neoadjuvant setting, prising nanoparticles comprising a rapamycin or a derivative 45 lung cancer (such as first line advanced NSCLC and thereof and a carrier protein (e.g., albumin), and b) an effec NSCLC), ovarian cancer, and melanoma (including meta tive amount of an anthracycline antibiotic. In some embodi static melanoma). ments, the invention provides a method of treating cancer in Many anti-angiogenic agents have been identified and are an individual, comprising administering to the individual a) known in the art, including those listed by Carmeliet and Jain an effective amount of a composition comprising nanopar 50 (2000). The anti-angiogenic agent can be naturally occurring ticles comprising rapamycin and an albumin, and b) an effec or non-naturally occurring. In some embodiments, the che tive amount of an anthracycline antibiotic. Suitable anthracy motherapeutic agent is a synthetic antiangiogenic peptide. cline antibiotic include, but are not limited to, Doxil (R), For example, it has been previously reported that the antian actinomycin, , (daunomycin), giogenic activity of Small synthetic pro-apoptic peptides doxorubicin (adriamycin), epirubicin, , mitox 55 comprise two functional domains, one targeting the CD13 antrone, and . In some embodiments, the anthracy receptors (aminopeptidase N) on tumor microVessels and the cline is any of (and in some embodiments selected from the other disrupting the mitochondrial membrane following group consisting of) DOXil.R., epirubicin, and doxorubicin. In internalization. Nat. Med. 1999, 5(9): 1032-8. A second gen some embodiments, the method is for treatment of an early eration dimeric peptide, CNGRC-GG-d (KLAKLAK)2. stage breast cancer. In some embodiments, the method is for 60 named HKP (Hunter Killer Peptide) was found to have treatment of a breast cancer in an adjuvant or a neoadjuvant improved antitumor activity. Accordingly, in some embodi Setting. ments, the antiangiogenic peptide is HKP. In some embodi In some embodiments, the invention provides a method of ments, the antiangiogenic agent is other than an anti-VEGF treating cancer in an individual, comprising administering to antibody (Such as Avastin R). In some embodiments, the anti the individuala) an effective amount of a composition com 65 angiogenic agent is a small molecule inhibitor of VEGFR prising nanoparticles comprising a rapamycin or a derivative (such as VEGFR1, VEGFR2, and/or VEGFR3). Suitable thereof and a carrier protein (e.g., albumin), and b) an effec small molecule inhibitors of VEGFR include, but are not US 8,911,786 B2 31 32 limited to, , AZD2171, pazopanib (GW786034), nanoparticle in the composition is administered by intrave Sunitinib, AG013736, Sorafenib, ZD6474, XL647, and nous administration. In some embodiments, the anti-VEGF XL999. antibody is administered by intravenous administration. In In some embodiments, the invention provides a method of Some embodiments, both the rapamycin or a derivative treating cancer in an individual, comprising administering to 5 thereof in the nanoparticle composition and the anti-VEGF the individuala) an effective amount of a composition com antibody are administered by intravenous administration. prising nanoparticles comprising a rapamycin or a derivative In some embodiments, there is provided a method of inhib thereof and a carrier protein (e.g., albumin), and b) an effec iting tumor metastasis in an individual, comprising adminis tive amount of a proteasome inhibitor, such as bortezomib tering to the individual: a) an effective amount of a composi (Velcade). In some embodiments, the invention provides a 10 tion comprising nanoparticles comprising rapamycin or a method of treating cancer in an individual, comprising administering to the individual a) an effective amount of a derivative thereof and a carrier protein (e.g., albumin), and b) composition comprising nanoparticles comprising rapamy an effective amount of an anti-VEGF antibody. In some cin and an albumin, and b) an effective amount of a protea embodiments, the effective amounts of the rapamycin or a some inhibitor such as bortezomib (Velcade). 15 derivative thereof nanoparticle composition and the anti In some embodiments, the invention provides a method of VEGF antibody synergistically inhibit tumor metastasis. In treating cancer in an individual, comprising administering to some embodiments, at least about 10% (including for the individuala) an effective amount of a composition com example at least about any of about 20%, 30%, 40%. 60%, prising nanoparticles comprising a rapamycin or a derivative 70%, 80%, 90%, or 100%) metastasis is inhibited. In some thereof and a carrier protein (e.g., albumin), and b) an effec embodiments, method of inhibiting metastasis to lymph node tive amount of a therapeutic antibody. In some embodiments, is provided. In some embodiments, method of inhibiting the invention provides a method of treating cancer in an metastasis to the lung is provided. In some embodiments, the individual, comprising administering to the individual a) an rapamycin or a derivative thereof is rapamycin. In some effective amount of a composition comprising nanoparticles embodiments, the anti-VEGF antibody is bevacizumab (such comprising rapamycin and an albumin, and b) an effective 25 as Avastin R). In some embodiments, the rapamycin or a amount of a therapeutic antibody. Suitable therapeutic anti derivative thereof in the nanoparticle in the composition is bodies include, but are not limited to, anti-VEGF antibody administered by intravenous administration. In some embodi (such as Avastin R) (bevacizumab)), anti-HER2 antibody ments, the anti-VEGF antibody is administered by intrave (such as HerceptinR) (trastuzumab)), Erbitux(R) (cetuximab), nous administration. In some embodiments, both the rapamy Campath (alemtuzumab), Myelotarg (gemtuzumab), Zevalin 30 cin or a derivative thereof in the nanoparticle composition and (ibritumomab tiuextan, Rituxan (rituximab), and Bexxar (to situmomab)). In some embodiments, the chemotherapeu the anti-VEGF antibody are administered by intravenous tic agent is ErbituXR (cetuximab). In some embodiments, the administration. chemotherapeutic agent is a therapeutic antibody other than In Some embodiments, two or more chemotherapeutic an antibody against VEGF or HER2. In some embodiments, 35 agents are administered in addition to the rapamycin or a the method is for treatment of HER2 positive breast cancer, derivative thereof in the nanoparticle composition. These two including treatment of advanced breast cancer, treatment of or more chemotherapeutic agents may (but not necessarily) metastatic cancer, treatment of breast cancer in an adjuvant belong to different classes of chemotherapeutic agents. setting, and treatment of cancer in a neoadjuvant setting. In Examples of these combinations are provided herein. Other some embodiments, the method is for treatment of any of 40 combinations are also contemplated. metastatic breast cancer, breast cancer in an adjuvant setting In some embodiments, there is provided a method of treat or a neoadjuvant setting, lung cancer (such as first line ing cancer in an individual, comprising administering to the advanced NSCLC and NSCLC), ovarian cancer, head and individuala) an effective amount of a composition compris neck cancer, and melanoma (including metastatic mela ing nanoparticles comprising a rapamycin or a derivative noma). For example, in Some embodiments, there is provided 45 thereof and a carrier protein (e.g., albumin), b) an effective a method for treatment of HER2 positive metastatic breast amount of an antimetabolite (such as a nucleoside analog, for cancer in an individual, comprising administering to the indi example, gemcitabine), and c) an anthracycline antibiotic vidual about 54 mg to 540 mg rapamycin or about 30 mg/m2 (such as epirubicin). In some embodiments, there is provided to 300 mg/m2 rapamycin in a nanoparticle composition a method of treating cancer in an individual, comprising weekly for three weeks with the fourth week off, concurrent 50 administering to the individual a) an effective amount of a with the administration of Herceptin R. composition comprising nanoparticles comprising rapamy In some embodiments, there is provided a method of treat cin and an albumin, b) an effective amount of an antimetabo ing cancer in an individual, comprising administering to the lite (such as a nucleoside analog, for example, gemcitabine), individual: a) an effective amount of a composition compris and c) an effective amount of an anthracycline antibiotic ing nanoparticles comprising rapamycin or a derivative 55 (such as epirubicin). In some embodiments, the method is for thereof and a carrier protein (e.g., albumin), and b) an effec treatment of breast cancer in a neoadjuvant setting. For tive amount of an anti-VEGF antibody. In some embodi example, in some embodiments, there is provided a method of ments, the effective amounts of the rapamycin or a derivative treating locally advanced/inflammatory cancer in an indi thereof nanoparticle composition and the anti-VEGF anti vidual comprising administering to the individual rapamycin body synergistically inhibit cell proliferation (such as tumor 60 (such as about 30 mg/m2 to about 300 mg/m2 or such as about cell growth). In some embodiments, at least about 10% (in 50 mg to 540 mg rapamycin) in a nanoparticle composition cluding for example at least about any of about 20%, 30%, every two weeks; 2000 mg/m2 gemcitabine, every two 40%, 60%, 70%, 80%, 90%, or 100%) cell proliferation is weeks; and 50 mg/m2 epirubicin, every two weeks. In some inhibited. In some embodiments, the rapamycin or a deriva embodiments, there is provided a method of treating breast tive thereof is rapamycin. In some embodiments, the anti 65 cancer in an individual in an adjuvant setting, comprising VEGF antibody is bevacizumab (such as AvastinR). In some administering to the individual rapamycin (such as about 30 embodiments, the rapamycin or a derivative thereof in the mg/m2 to about 300 mg/m2 or such as about 50 mg to 540 mg US 8,911,786 B2 33 34 rapamycin) in a nanoparticle composition every two weeks, adriamycin, and 600 mg/m2 cyclophosphamide, wherein the 2000 mg/m2 gemcitabine, every two weeks, and 50 mg/m2 administration is carried out once every two weeks. epirubicin, every two weeks. In some embodiments, the invention provides a method of In some embodiments, there is provided a method of treat treating cancer in an individual, comprising administering to ing cancer in an individual, comprising administering to the the individuala) an effective amount of a composition com individuala) an effective amount of a composition compris prising nanoparticles comprising a rapamycin or a derivative ing nanoparticles comprising a rapamycin or a derivative thereof and a carrier protein (e.g., albumin)and b) an effective thereof and a carrier protein (e.g., albumin), b) an effective amount of an p110C.-specific inhibitor (e.g., PX-866). In amount of a platinum-based agent (such as carboplatin), and Some embodiments, the method further comprises adminis c) a therapeutic antibody (such as ant-HER2 antibody (such 10 tering an effective amount of a tyrosine kinase inhibitor (e.g., gefitinib or erlotinib). In some embodiments, the cancer is as Herceptin R) and anti-VEGF antibody (such as AvastinR)). non-Small cell lung carcinoma. In some embodiments, there is provided a method of treating In some embodiments, the invention provides a method of cancer in an individual, comprising administering to the indi treating cancer in an individual, comprising administering to vidual a) an effective amount of a composition comprising 15 the individuala) an effective amount of a composition com nanoparticles comprising rapamycin and an albumin, b) an prising nanoparticles comprising a rapamycin or a derivative effective amount of a platinum-based agent (Such as carbopl thereof and a carrier protein (e.g., albumin)and b) an effective atin), and c) a therapeutic antibody (such as ant-HER2 anti amount of a compound that affects the MAPK pathway (e.g., body (such as Herceptin R) and anti-VEGF antibody (such as Sorafenib (BAY49-9006). In some embodiments, the method Avastin R)). In some embodiments, the method is for treat further comprises administering an effective amount of a ment of any of advanced breast cancer, metastatic breast tyrosine kinase inhibitor (e.g., gefitinib or erlotinib). In some cancer, breast cancer in an adjuvant setting, and lung cancer embodiments, the cancer is non-Small cell lung carcinoma. In (including NSCLC and advanced NSCLC). In some embodi Some embodiments, the cancer is brain cancer (e.g., glioblas ments, there is provided a method of treating metastatic can toma). cer in an individual, comprising administering to the indi 25 In some embodiments, there is provided a method of treat vidual rapamycin (such as about 30 mg/m2 to about 300 ing cancer in an individual, comprising administering to the mg/m2 or such as about 50 mg to 540 mg rapamycin) in a individuala) an effective amount of a composition compris nanoparticle composition and carboplatin, AUC-2, wherein ing nanoparticles comprising a rapamycin or a derivative the administration is carried out weekly for three weeks with thereof and a carrier protein (e.g., albumin)and b) an effective the fourth week off. In some embodiments, the method fur 30 amount of another agent that affects a signaling pathway ther comprises weekly administering about 2-4 mg/kg of involving a target of rapamycin. In some embodiments, there Herception(R). is provided a method of treating cancer in an individual, In some embodiments, there is provided a method of treat comprising administering to the individual a) an effective ing cancer in an individual, comprising administering to the amount of a composition comprising nanoparticles compris individuala) an effective amount of a composition compris 35 ing a rapamycin or a derivative thereof and a carrier protein ing nanoparticles comprising a rapamycin or a derivative (e.g., albumin) and b) an effective amount of another agent thereof and a carrier protein (e.g., albumin), b) an effective that affects a signaling pathway involving mTOR. In some amount of a platinum-based agent (such as carboplatin), and embodiments, the other agent affects a signaling pathway c) a Vinca alkaloid (such as Navelbine(R). In some embodi involving TORC1. In some embodiments, the other agent ments, there is provided a method of treating cancer in an 40 affects a signaling pathway involving mTORC2. Signaling individual, comprising administering to the individual a) an pathways involving mTOR include, but are not limited to, effective amount of a composition comprising nanoparticles PI3K/Akt pathwayn and cAMP/AMPK pathway. These path comprising rapamycin and an albumin, b) an effective ways are interrelated. Accordingly, an agent that affects one amount of a platinum-based agent (such as carboplatin), and signaling pathway frequently affects the other pathway (ei c) a Vinca alkaloid (such as Navelbine(R). In some embodi 45 ther directly or indirectly). ments, the method is for treatment of lung cancer. In some embodiments, the signaling pathway involving In some embodiments, the invention provides a method of mTOR is the PI3K/Akt signaling pathway. For example, in treating cancer in an individual, comprising administering to Some embodiments, there is provided a method of treating the individuala) an effective amount of a composition com cancer in an individual, comprising administering to the indi prising nanoparticles comprising a rapamycin or a derivative 50 vidual a) an effective amount of a composition comprising thereof and a carrier protein (e.g., albumin), b) an effective nanoparticles comprising a rapamycin or a derivative thereof amount of an alkylating agent (Such as cyclophosphamide) and a carrier protein (e.g., albumin) and b) an effective and c) an anthracycline antibiotic (such as adriamycin). In amount of another agent that inhibits PI3K/Akt activation. In Some embodiments, the invention provides a method of treat some embodiments, the cancer is any of HER2+ breast can ing cancer in an individual, comprising administering to the 55 cer, chronic mylogenous leukemia CML, ovarian cancer, individuala) an effective amount of a composition compris endometrial cancer, sarcoma, SCCHN (squamous cell carci ing nanoparticles comprising rapamycin and an albumin, b) nomaterm of the head and neck), and thyroid cancer. an effective amount of an alkylating agent (Such as cyclo The PI3/Akt signaling pathway described herein includes phosphamide) and c) an anthracycline antibiotic (such as any members or components that directly or indirectly par adriamycin). In some embodiments, the method is for treat 60 ticipate in the signal transduction cascade. These include, but ment of an early stage breast cancer. In some embodiments, are not limited to, PI3 kinase, Akt, PDK1, RAPTOR (regula the method is for treatmentofa breast cancer in an adjuvant or tory associated protein if mTOR), TSC1 (tuberous sclerosis a neoadjuvant setting. For example, in some embodiments, complex 1), TSC2, PTEN (phosphatase and tenesin there is provided a method of treating an early stage breast homolog), and downstream effectors such as cyclin D, HIF1, cancer in an individual, comprising administering rapamycin 65 HIF2, Glut1, LAT1, and c-Myc. Components of the PI3/Akt (such as about 30 mg/m2 to about 300 mg/m2 or 50 mg to 540 signaling pathway may also include RHEB, Rictor, S6K, mg rapamycin) in a nanoparticle composition, 60 mg/m2 4EBP1, cAMP. cAMPK, GBL. IRS, PIP2, PIP3, Rho, Ras, US 8,911,786 B2 35 36 Abl, PKC. eIF4E, PDGFR, VEGFR, and VHL. The agent that While the agents described herein are sometimes referred affects (such as inhibits) the PI3K/Akt signaling pathway can to as signaling pathway inhibitors, the methods described thus act through modulation of any one or more of these herein includes the use of these inhibitors to treat cancer components. regardless of the mechanism of action or how the therapeutic In some embodiments, the other agent inhibits PI3 kinase effect is achieved. Indeed, it is recognized that Such com (PI3K). Suitable inhibitors of PI3K include, but are not lim pounds may have more than one target, and the initial activity ited to, wortmannin and the derivatives or analogs thereof. recognized for a compound may not be the activity that it and analogs thereof, such as OSU-03012 and OSU possesses in vivo when administered to a subject, or whereby 03013: 3-deoxy-D-myo-inositol analogs, such as PX-316; it achieves its therapeutic efficacy. Thus, the description of a 2-substituted 3'-deoxy-phosphatidyl-myo-inositol analogs; 10 compound as a pathway or protein target (e.g., Akt or mTOR) fused heteroaryl derivatives; 3-(imidazo 1,2-alpyridin-3-yl) inhibitor indicates that a compound possesses such activity, derivatives: Ly294.002; quinazoline-4-one derivatives, such but in no way restricts a compound to having that activity as IC486068; 3-(hetero)aryloxy substituted benzo(b) when used as a therapeutic or prophylactic agent. thiophene derivatives; viridins, including semi-synthetic viri 15 Other agents that can be used in combination with rapamy dins such as such as PX-866 (acetic acid (1S, 4E. 10R, 11 R, cin (or its derivative) compositions described herein include, 13S, 14R)-4-dially laminomethylene-6-hydroxy-1-meth for example, flavopiridol, , SN38, inhibitor of oxymethyl-10,13-dimethyl-3,7,17-trioxo-1,3,4,7,10,11,12. breast cancer resistant protein (such as KO143 and fumitrem 13,14, 15,16,17-dodecahydro-2-Oxa-cyclopentaaphenan orgin C). thren-11-yl ester); and wortmannin and derivatives thereof. In some embodiments, there is provided a method of treat In some embodiments, the other agent inhibits Akt kinase, ing advanced breast cancer in an individual, comprising including Akt1, Akt2, and Akt3. In some embodiments, the administering to the individual a) an effective amount of a other agent inhibits phosphorylation of 5473 of the human composition comprising nanoparticles comprising a rapamy Akt kinase, but not T308. In some embodiments, the second cin and an albumin, b) an effective amount of carboplatin. In compound inhibits phosphorylation of T308 of the human 25 Some embodiments, the method further comprises adminis Akt kinase, but not S473. In some embodiments, the other tering an effective amount of Herceptin R) to the individual. In agent inhibits phorphorylation of both S473 and T308 of the Some embodiments, there is provided a method of treating Akt kinase. In some embodiments, the other agent interferes metastatic breast cancer in an individual, comprising admin with the membrane localization of the Akt kinase. Suitable istering to the individual a) an effective amount of a compo inhibitors of Akt kinase include, but are not limited to, Akt 30 sition comprising nanoparticles comprising rapamycin and 1-1 (inhibits Akt 1), Akt-1-1.2 (inhibits Akt1 and 2), API an albumin, b) an effective amount of gemcitabine. In some 59CJ-Ome. 1-H-imidazo[4,5-cpyridinyl compounds, embodiments, there is provided a method of treating indole-3-carbinol and derivatives thereof, perifosine, phos advanced non-Small cell lung cancer in an individual, com phatidylinositol ether lipid analogues, triciribine (TCN or prising administering to the individuala) an effective amount API-2 or NCI identifier: NSC 154020). In some embodi 35 of a composition comprising nanoparticles comprising rapa ments, the other agent is perifosine. mycin and an albumin, b) an effective amount of carboplatin. In some embodiments, the other agent is an inhibitor of In some embodiments, the method further comprises iden PDK1. tifying a cancer patient population (e.g., breast cancer) based In some embodiments, there is provided a method of treat on a hormone receptor status of patients having tumor tissue ing cancer in an individual, comprising administering to the 40 not expressing both ER and PgR and administering to the individuala) an effective amount of a composition compris patient population an effective amount of a composition com ing nanoparticles comprising a rapamycin or a derivative prising nanoparticles comprising rapamycin or a derivative thereofanda carrier protein (e.g., albumin) and b) an effective thereof and a carrier protein (e.g., albumin). In some embodi amount of another agent that inhibits cyclin D1 (Such as ments, the method further comprises administering to the cycline D1 overexpression). In some embodiments, the can 45 patient population an effective amount of at least one other cer is any of mantle cell lymphoma and breast cancer. chemotherapeutic agent. The at least one other chemothera In some embodiments, there is provided a method of treat peutic agent may be administered concurrently or sequen ing cancer in an individual, comprising administering to the tially with rapamycin or a derivative thereof nanoparticles. In individuala) an effective amount of a composition compris Some embodiments, the at least one other chemotherapeutic ing nanoparticles comprising a rapamycin or a derivative 50 agent comprises 5-Fluoruracil, Epirubicin and Cyclophos thereofanda carrier protein (e.g., albumin) and b) an effective phamide (FEC) administered concurrently or sequentially. amount of another agent that inhibits Mycover expression. In These methods may have higher efficacy in ER(-)/PgR(-) Some embodiments, the cancer is burkitt lymphoma. populations in all patient populations, both HER-2 positive In some embodiments, the other agent inhibits HIF. In and HER-2 negative. some embodiments, the HIF is HIF1. In some embodiments, 55 In some embodiments of any of the above methods of the HIF is HIF2. In some embodiments, there is provided a combination therapy with a chemotherapeutic agent, there is method of treating cancer in an individual, comprising provided a composition comprising nanoparticles compris administering to the individual a) an effective amount of a ing rapamycin or a derivative thereof and a carrier protein composition comprising nanoparticles comprising a rapamy (such as albumin) and at least one other chemotherapeutic cin or a derivative thereof and a carrier protein (e.g., albumin) 60 agent. The compositions described herein may comprise and b) an effective amount of another agent that inhibits HIF effective amounts of the rapamycin or a derivative thereofand (such as HIF overexpression). In some embodiments, the the chemotherapeutic agent for the treatment of a cancer. In other agent inhibits HIF-mediated angiogenesis. In some Some embodiments, the chemotherapeutic agent and rapamy embodiments, the cancer is RCC and Von Hippel-Lindau cin or a derivative thereof are present in the composition at a (VHL). 65 predetermined ratio, such as the weight ratios described Other PI3K/Akt signaling pathway inhibitors include, but herein. In some embodiments, the invention provides a syn are not limited to, e.g., FTY720 and UCN-01. ergistic composition of an effective amount of a composition US 8,911,786 B2 37 38 comprising nanoparticles comprising rapamycin or a deriva motherapeutic agents. For example, the individual may first tive thereof and an effective amount of at least one other be administered with a rapamycin or its derivative thereof. chemotherapeutic agent. containing nanoparticle composition and at least one other In some embodiments of any of the above methods of chemotherapeutic agent, and Subsequently be subject to hor combination therapy with a chemotherapeutic agent, the mone therapy, radiation therapy, and/or Surgery. Alterna invention provides pharmaceutical compositions comprising tively, the individual may first be treated with hormone nanoparticles comprising a rapamycin or a derivative thereof therapy, radiation therapy, and/or Surgery, which is then fol and a carrier protein (such as albumin) for use in the treatment lowed by the administration of a nanoparticle composition of a cancer, wherein said use comprises simultaneous and/or and at least one other chemotherapeutic agent. Other combi sequential administration of at least one other chemothera 10 nations are also contemplated. peutic agent. In some embodiments, the invention provides a Administration of nanoparticle compositions disclosed pharmaceutical composition comprising a chemotherapeutic above in conjunction with administration of chemotherapeu agent for use in the treatment of a cancer, wherein said use tic agent is equally applicable to those in conjunction with comprises simultaneous and/or sequential administration of a hormone therapy, radiation therapy, and/or Surgery. composition comprising nanoparticles comprising rapamy 15 The term hormonetherapy, as used herein, includes, but is cin or a derivative thereof and a carrier protein (such as not limited to, androgen ablation therapy, androgen depriva albumin). In some embodiments, the invention provides rapa tion therapy, hormonal ablation therapy, combined hormone mycin or a derivative thereof-containing nanoparticle com blockade, intermittent hormonal therapy, neoadjuvant hor positions and compositions comprising one other chemo monal therapy, neoadjuvant androgen Suppression, and neo therapeutic agent for simultaneous, and/or sequential use for adjuvantandrogen deprivation. Androgens, such as testoster treatment of a cancer. one, regulate the growth, differentiation, and rate of apoptosis In some embodiments, the invention provides a method to in the prostate and its malignancies. In some embodiments, treat cancer comprising administering to an individual an prostate cancer may be treated by exploiting the general effective amount of a composition comprising nanoparticles dependency of prostate cancer on androgen through several comprising a rapamycin or a derivative thereof and a carrier 25 therapies referred to as hormonetherapy. protein (Such as albumin) simultaneously and/or sequentially In some embodiments, there is provided a method of treat with Surgery, radiation, gene therapy, immunotherapy, bone ing cancer in an individual, comprising administering to the marrow transplantation, stem cell transplantation, hormone individuala) an effective amount of a composition compris therapy, targeted therapy, cryotherapy, ultrasound therapy, ing nanoparticles comprising a rapamycin or a derivative and/or photodynamic therapy. In some embodiments, the 30 thereof and a carrier protein (e.g., albumin)and b) an effective present invention provides a method of treating cancer com amount of a gonadotropin-releasing hormone (GnRH) ago prising a first therapy comprising administering nanoparticles nist (also called LHRH agonist, luteinizing-hormone releas comprising rapamycin and an albumin, and a second therapy ing hormone agonist). In some embodiments, there is pro comprising Surgery, radiation, gene therapy, immunotherapy, vided a method of treating cancer in an individual, bone marrow transplantation, stem cell transplantation, hor 35 comprising administering to the individual a) an effective mone therapy, targeted therapy, cryotherapy, ultrasound amount of a composition comprising nanoparticles compris therapy, and/or photodynamic therapy. In some embodi ing rapamycin and an albumin and b) an effective amount of ments, the cancer may be prostate cancer. In some embodi a GnRH agonist. In some embodiments, the method is for ments, the second therapy is hormone therapy. In some treatment of prostate cancer. In some embodiments, the embodiments, the second therapy is radiation therapy. In 40 invention provides pharmaceutical compositions comprising Some embodiments, the second therapy is Surgery. nanoparticles comprising a rapamycin or a derivative thereof The administration of rapamycin or a derivative thereof and a carrier protein (such as albumin) for use in the treatment nanoparticle composition may be prior to the hormone of a cancer, wherein said use comprises simultaneous and/or therapy, radiation, and/or Surgery, after the hormone therapy, sequential administration of at least one GnRHagonist. Suit radiation, and/or Surgery, or concurrent with hormone 45 able therapeutic GnRH agonists include, but are not limited therapy, radiation, and/or Surgery. For example, the adminis to, leuprolide, goserelin, naferelin, meterelin, buserelin, his tration of rapamycin or a derivative thereofnanoparticle com torelin, deslorelin, and triptorelin. position may precede or follow hormone therapy, radiation, In some embodiments, there is provided a method of treat and/or Surgery therapy by intervals ranging from minutes to ing cancer in an individual, comprising administering to the weeks. In some embodiments, the time period between the 50 individuala) an effective amount of a composition compris first and the second therapy is such that the rapamycin or a ing nanoparticles comprising a rapamycin or a derivative derivative thereof and a carrier protein (e.g., albumin) and thereof and a carrier protein (e.g., albumin), b) an effective hormonetherapy, radiation, and/or Surgery would still be able amount of a GnRH agonist, and c) antiandrogen. In some to exert an advantageously combined effect on the cell. In embodiments, there is provided a method of treating cancer in some embodiments, it may be desirable to extend the time 55 an individual, comprising administering to the individual a) period for treatment significantly, where several days to sev an effective amount of a composition comprising nanopar eral weeks lapse between the two therapies. ticles comprising rapamycin and an albumin, b) an effective Surgery described herein includes resection in which all or amount of a GnRH agonist, c) and an antiandrogen. In some part of cancerous tissue is physically removed, exercised, embodiments, the method is for treatment of prostate cancer. and/or destroyed. Tumor resection refers to physical removal 60 In some embodiments, the antiandrogen administration of at least part of a tumor. In addition to tumor resection, begins prior to treatment with the GnRH agonist and/or the treatment by Surgery includes laser Surgery, cryoSurgery, rapamycin-containing nanoparticle composition. In some electroSurgery, and micropically controlled Surgery (Mohs embodiments, the invention provides pharmaceutical compo Surgery). Removal of Superficial Surgery, precancers, or nor sitions comprising nanoparticles comprising a rapamycin or a mal tissues are also contemplated. 65 derivative thereof and a carrier protein (such as albumin) for The hormone therapy, radiation therapy, and/or Surgery use in the treatment of a cancer, wherein said use comprises may be carried out in addition to the administration of che simultaneous and/or sequential administration of at least one US 8,911,786 B2 39 40 GnRH agonist or antiandrogen. In some embodiments, the blocker, inhibitor, or antagonist. In some embodiments, the antiandrogenis administered before the GnRHagonist and/or method is for treatment of prostate cancer (such as advanced the rapamycin-containing nanoparticle composition, and the prostate cancer). In some embodiments, the invention pro administration of the antiandrogen is continued for at least the vides pharmaceutical compositions comprising nanoparticles first month of GnRH agonist therapy. In some embodiments, comprising a rapamycin or a derivative thereof and a carrier the antiandrogen administration begins any of about 1, 2, 3, 4, protein (Such as albumin) for use in the treatment of a cancer, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, wherein said use comprises simultaneous and/or sequential 23, and 24 weeks prior to treatment with the GnRH agonist administration of at least one endothelin-A receptor blocker, and/or the rapamycin-containing nanoparticle composition inhibitor, or antagonist. A Suitable therapeutic endothelin-A Suitable therapeutic GnRH agonists include, but are not lim 10 receptor blocker, inhibitor, or antagonist includes, but is not ited to, leuprolide, goserelin, naferelin, meterelin, buserelin, limited to, (ABT 627, Abbott Laboratories, historelin, deslorelin, and triptorelin. Suitable therapeutic Abbott Park, Ill.). antiandrogens include, but are not limited to, bicalutamide It is understood that any of the methods of treating cancer (Casodex), flutamide (Eulexin), cyproterone, nilutamide (Ni described herein (such as above section “Methods of Treating landron), and other therapeutic agents that are effective in 15 Cancer) apply to and include description of combination ultimately reducing circulating androgen levels to the castrate therapies. In some embodiments of any of the methods of level treatment related to combination therapy described herein, In some embodiments, there is provided a method of treat treatment with the combination of the first therapy (e.g., a ing cancer in an individual, comprising administering to the nanoparticle composition comprising rapamycin or a deriva individuala) an effective amount of a composition compris tive thereofanda carrier protein) and the second therapy (e.g., ing nanoparticles comprising a rapamycin or a derivative one or more compounds useful for treating cancer) may result thereofanda carrier protein (e.g., albumin) and b) an effective in an additive or even synergistic (e.g., greater than additive) amount of a gonadotropin-releasing hormone (GnRH) result compared to administration of either therapy alone. In antagonist (also called LHRH antagonist, luteinizing-hor Some embodiments, a lower amount of each pharmaceuti mone releasing hormone antagonist). In some embodiments, 25 cally active compound is used as part of a combination there is provided a method of treating cancer in an individual, therapy compared to the amount generally used for individual comprising administering to the individual a) an effective therapy. Preferably, the same or greater therapeutic benefit is amount of a composition comprising nanoparticles compris achieved using a combination therapy than by using any of the ing rapamycin and an albumin and b) an effective amount of individual compounds alone. In some embodiments, the same a GnRH antagonist. In some embodiments, the method is for 30 or greater therapeutic benefit is achieved using a smaller treatment of prostate cancer. In some embodiments, the amount (e.g., a lower dose or a less frequent dosing schedule) invention provides pharmaceutical compositions comprising of a pharmaceutically active compound in a combination nanoparticles comprising a rapamycin or a derivative thereof therapy than the amount generally used for individual and a carrier protein (such as albumin) for use in the treatment therapy. Preferably, the use of a small amount of pharmaceu of a cancer, wherein said use comprises simultaneous and/or 35 tically active compound results in a reduction in the number, sequential administration of at least one GnRH antagonist. severity, frequency, or duration of one or more side-effects Suitable therapeutic GnRH antagonist include, but are not associated with the compound. limited to, Cetrorelix acetate (Cetrotide), by Serono, Ganire In some embodiments of any of the methods of treatment lix acetate (Antagon), by Organon International, Abarelix related to combination therapy, the rapamycin or derivative (Plenaxis), and the like. 40 thereof and the second compound (e.g., a chemotherapeutic In an embodiment, the method comprises the administra agent and/or hormone therapeutic agent) are present in a tion of a therapeutic effective amount of a rapamycin-con single composition containing at least two different nanopar taining nanoparticle composition at any one or more of the ticles, wherein some of the nanoparticles in the composition following times: prior to hormone therapy, in conjunction comprise the rapamycin or derivative thereof and a carrier with hormonetherapy, during hormonetherapy, or following 45 protein, and some of the other nanoparticles in the composi hormone therapy to treat prostate cancer. In some embodi tion comprise a second pharmaceutically active compound ments, the method comprises the administration of a thera and a carrier protein. In some embodiments, only the rapa peutic effective amount of a rapamycin-containing nanopar mycin or derivative thereof is contained in nanoparticles. In ticle composition either simultaneously with or separately Some embodiments, simultaneous administration of rapamy from the hormonetherapeutic agent to treat prostate cancer. A 50 cin or derivative thereof in the nanoparticle composition and combination of a therapeutically effective amount of one or the second compound can be combined with Supplemental more standard hormone therapy drugs and a therapeutically doses of rapamycin and/or the second compound. effective amount of rapamycin or a derivative in a nanopar In some embodiments of any of the above embodiments ticle composition may result in a synergistic effect in prostate related to combination therapies described herein, the first tumor inhibition (including regression of existing prostate 55 and second therapies are administered simultaneously, either tumor). in the same composition or in separate compositions. In some In some embodiments, there is provided a method of treat embodiments, the first and second therapies are administered ing cancer in an individual, comprising administering to the sequentially, i.e., the first therapy is administered either prior individuala) an effective amount of a composition compris to or after the administration of the first and second therapy. In ing nanoparticles comprising a rapamycin or a derivative 60 Some embodiments, the administration of the first and second thereofanda carrier protein (e.g., albumin) and b) an effective therapies is concurrent, i.e., the administration period of the amount of an endothelin-A receptor blocker, inhibitor, or first therapy and that of the second therapy overlap with each antagonist. In some embodiments, there is provided a method other. In some embodiments, the administration of the first of treating cancer in an individual, comprising administering and second therapies is non-concurrent. For example, in some to the individual a) an effective amount of a composition 65 embodiments, the administration of the first therapy is termi comprising nanoparticles comprising rapamycin and an albu nated before the second therapy is administered. In some min and b) an effective amount of an endothelin-A receptor embodiments, the administration of the second therapy is US 8,911,786 B2 41 42 terminated before the first therapy is administered. In some In Some embodiments, the amount of rapamycin or a embodiments, the second therapy is radiation therapy. In derivative thereof in the composition is an amount sufficient Some embodiments, the second therapy is Surgery. to increase basal AKT activity, increase AKT phosphoryla In some embodiments of any of the above embodiments tion, increase PI3-kinase activity, increase the length of acti related to combination therapy, the cancer is early stage can Vation of AKT (e.g., activation induced by exogenous IGF-1), cer, non-metastatic cancer, primary cancer, advanced cancer, inhibit serine phosphorylation of IRS-1, inhibit IRS-1 degra locally advanced cancer, metastatic cancer, cancer in remis dation, inhibit or alter CXCR4 subcellular localization, Sion, recurrent cancer, cancer in an adjuvant setting, cancer in inhibit VEGF secretion, decrease expression of cyclin D2, a neoadjuvant setting, or cancer Substantially refractory to decrease expression of survivin, inhibit IL-6-induced mul 10 tiple myeloma cell growth, inhibit cancer cell proliferation, hormone therapy. In some embodiments, the cancer is a Solid increase apoptosis, increase cell cycle arrest, increase cleav tumor. In some embodiments, the cancer is not a solid tumor age of poly(ADPribose) polymerase, increase cleavage of (i.e., other than a Solid tumor). In some embodiments, the caspase-8/caspase-9, alter or inhibit signaling in the phos cancer is a plasmacytoma. In some embodiments, the cancer phatidylinositol 3-kinase/AKT/mTOR and/or cyclin D1/ret is multiple myeloma, renal cell carcinoma, prostate cancer, 15 inoblastoma pathways, inhibit angiogenesis, and/or inhibit lung cancer, melanoma, brain cancer (e.g., glioblastoma). osteoclast formation. ovarian cancer, or breast cancer. In some embodiments, the In some embodiments, the invention provides a method of cancer is a carcinoma (i.e., other than a carcinoma). In some treating cancer in an individual by administering to the indi embodiments, the cancer is not colon cancer (i.e., other than vidual (e.g., a human) an effective amount of a composition colon cancer). In some embodiments, the cancer is not breast comprising nanoparticles that comprise rapamycin or a cancer (i.e., other than breast cancer). In some embodiments, derivative thereof and a carrier protein (e.g., albumin Such as the cancer is not ovarian cancer, prostate cancer, or brain human serum albumin). In some embodiments, the amount of CaCC. rapamycin or a derivative thereof in the composition is In some embodiments of any of the above embodiments included in any of the following ranges: about 0.5 to about 5 related to combination therapy, a taxane is not administered to 25 mg, about 5 to about 10 mg, about 10 to about 15 mg, about 15 the individual. In some embodiments, the taxane adminis to about 20 mg, about 20 to about 25 mg, about 20 to about 50 tered is not a nanoparticle composition. In some embodi mg, about 25 to about 50 mg, about 50 to about 75 mg, about ments, the nanoparticle composition comprising rapamycin 50 to about 100 mg, about 75 to about 100 mg, about 100 to or a derivative thereof is not administered in conjunction with about 125 mg, about 125 to about 150 mg, about 150 to about a taxane. In some embodiments, a taxane is not administered 30 175 mg, about 175 to about 200 mg, about 200 to about 225 to the individual during the time period in which the indi mg, about 225 to about 250 mg, about 250 to about 300 mg. vidual is receiving one or more doses of a nanoparticle com about 300 to about 350 mg, about 350 to about 400 mg, about position comprising rapamycin or a derivative thereof In 400 to about 450 mg. or about 450 to about 500 mg. In some Some embodiments, the individual was treated with a taxane embodiments, the amount of rapamycin or derivative thereof before treatment begins with a nanoparticle composition 35 in the effective amount of the composition (e.g., a unit dosage comprising rapamycin or a derivative thereof For example, form) is in the range of about 54 mg to about 540 mg. Such as the individual may have received a taxane one or more days, about 180 mg to about 270 mg or about 216 mg. In some weeks, months, or years before treatment begins with a nano embodiments, the concentration of the rapamycin in the com particle composition comprising rapamycin or a derivative position is dilute (about 0.1 mg/ml) or concentrated (about thereofIn other embodiments, the individual never receives a 40 100 mg/ml), including for example any of about 0.1 to about taxane before treatment begins with a nanoparticle composi 50 mg/ml, about 0.1 to about 20 mg/ml, about 1 to about 10 tion comprising rapamycin or a derivative thereof In some mg/ml, about 2 mg/ml to about 8 mg/ml, about 4 to about 6 embodiments, the individual is treated with a taxane after mg/ml, about 5 mg/ml. In some embodiments, the concen treatment with a nanoparticle composition comprising rapa tration of the rapamycin is at least about any of 0.5 mg/ml, 1.3 mycin or derivative thereof terminates. In other embodi 45 mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/ml. 4 mg/ml, 5 mg/ml, 6 ments, the individual is never treated with a taxane after mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 treatment with a nanoparticle composition comprising rapa mg/ml, 25 mg/ml, 30 mg/ml. 40 mg/ml, or 50 mg/ml. mycin or derivative thereof terminates. In some embodi Exemplary effective amounts of rapamycin or a derivative ments, the composition, first therapy, and/or second therapy thereof in the nanoparticle composition include, but are not do not contain a taxane. In other embodiments, the composi 50 limited to, about any of 25 mg/m, 30 mg/m, 50 mg/m, 60 tion, first therapy, and/or second therapy comprise a taxane. mg/m, 75 mg/m, 80 mg/m, 90 mg/m, 100 mg/m, 120 Dosing and Method of Administration mg/m, 160 mg/m, 175 mg/m, 180 mg/m,200 mg/m,210 The dose of the inventive composition administered to an mg/m, 220 mg/m,250 mg/m, 260 mg/m,300 mg/m,350 individual (Such as a human) may vary with the particular mg/m, 400 mg/m, 500 mg/m, 540 mg/m, 750 mg/m. composition, the method of administration, and the particular 55 1000 mg/m, or 1080 mg/m rapamycin. In various embodi stage of cancer being treated. The amount should be sufficient ments, the composition includes less than about any of 350 to produce a desirable response, such as a therapeutic or mg/m,300 mg/m,250 mg/m,200 mg/m, 150 mg/m, 120 prophylactic response against cancer. In some embodiments, mg/m, 100 mg/m, 90 mg/m, 50 mg/m, or 30 mg/m rapa the amount of the composition is a therapeutically effective mycin or a derivative thereof. In some embodiments, the amount. In some embodiments, that amount of the composi 60 amount of the rapamycin or a derivative thereof per adminis tion is a prophylactically effective amount. In some embodi tration is less than about any of 25 mg/m, 22 mg/m. 20 ments, the amount of rapamycin or a derivative thereof in the mg/m, 18 mg/m, 15 mg/m, 14 mg/m, 13 mg/m, 12 composition is below the level that induces a toxicological mg/m, 11 mg/m, 10 mg/m.9 mg/m, 8 mg/m, 7 mg/m, 6 effect (i.e., an effect above a clinically acceptable level of mg/m.5 mg/m, 4 mg/m.3 mg/m, 2 mg/m, or 1 mg/m. In toxicity) or is at a level where a potential side effect can be 65 Some embodiments, the effective amount of rapamycin or a controlled or tolerated when the composition is administered derivative thereof in the composition is included in any of the to the individual. following ranges: about 1 to about 5 mg/m, about 5 to about US 8,911,786 B2 43 44 10 mg/m, about 10 to about 25 mg/m, about 25 to about 50 week; and 150-250 mg/m twice a week. The dosing fre mg/m, about 50 to about 75 mg/m, about 75 to about 100 quency of the composition may be adjusted over the course of mg/m, about 100 to about 125 mg/m, about 125 to about the treatment based on the judgment of the administering 150 mg/m, about 150 to about 175 mg/m, about 175 to physician. about 200 mg/m, about 200 to about 225 mg/m, about 225 In some embodiments, the composition is administered to about 250 mg/m, about 250 to about 300 mg/m, about about 20 to about 40 mg/kg three times weekly. In some 300 to about 350 mg/m, or about 350 to about 400 mg/m. embodiments, the composition is administered about 60 to Preferably, the effective amount of rapamycin or a derivative about 120 mg/m, three times weekly or about 90 mg/m thereof in the composition is about 30 to about 300 mg/m. daily. In some embodiments, the composition is administered such as about 100 to about 150 mg/m, about 120 mg/m, 10 about 30 mg/kg daily. In some embodiments, methods of about 130 mg/m, or about 140 mg/m. treating multiple myeloma following these dosing regimes In some embodiments of any of the above aspects, the are provided. effective amount of rapamycin or a derivative thereof in the In yet another aspect, the invention provides a method of composition includes at least about any of 1 mg/kg, 2.5 treating cancer in an individual by parenterally administering mg/kg, 5 mg/kg, 7.5 mg/kg, 10 mg/kg, 15 mg/kg, or 20 15 to the individual (e.g., a human) an effective amount of a mg/kg. In various embodiments, the effective amount of rapa composition comprising nanoparticles that comprise rapa mycin or a derivative thereof in the composition includes less mycin or a derivative thereof and a carrier protein (e.g., albu than about any of 350 mg/kg, 300 mg/kg, 250 mg/kg, 200 min Such as human serum albumin). In some embodiments, mg/kg, 150 mg/kg, 100 mg/kg, 50 mg/kg, 25 mg/kg, 20 the route of administration is intravenous, intra-arterial, intra mg/kg, 10 mg/kg, 5 mg/kg, or 1 mg/kg rapamycin or a deriva muscular, or Subcutaneous. In various embodiments, about tive thereof. 54 mg to about 540 mg, such as about 180 mg to about 270 mg Exemplary dosing frequencies include, but are not limited or about 216 mg. of the rapamycin or derivative thereof is to, weekly without break; weekly, three out of four weeks: administered per dose. In some embodiments, a taxane is not once every three weeks; once every two weeks; weekly, two contained in the composition. In some embodiments, the out of three weeks. In some embodiments, the composition is 25 rapamycin or derivative thereof is the only pharmaceutically administered about once every 2 weeks, once every 3 weeks, active agent for the treatment of cancer that is contained in the once every 4 weeks, once every 6 weeks, or once every 8 composition. weeks. In some embodiments, the composition is adminis The compositions described herein can be administered to tered at least about any of 1x, 2x, 3x, 4x, 5x, 6.x, or 7x (i.e., an individual (such as human) via various routes, including, daily) a week. In some embodiments, the intervals between 30 for example, intravenous, intra-arterial, intraperitoneal, intra each administration are less than about any of 6 months, 3 pulmonary, oral, inhalation, intravesicular, intramuscular, months, 1 month, 20 days, 15, days, 12 days, 10 days, 9 days, intra-tracheal, subcutaneous, intraocular, intrathecal, trans 8 days, 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. mucosal, and transdermal. In some embodiments, Sustained In some embodiments, the intervals between each adminis continuous release formulation of the composition may be tration are more than about any of 1 month, 2 months, 3 35 used. For example, the inventive composition can be admin months, 4 months, 5 months, 6 months, 8 months, or 12 istered by inhalation to treat conditions of the respiratory months. In some embodiments, there is no break in the dosing tract. The composition can be used to treat respiratory condi schedule. In some embodiments, the interval between each tions such as pulmonary fibrosis, broncheolitis obliterans, administration is no more than about a week. lung cancer, bronchoalveolar carcinoma, and the like. In one The administration of the composition can be extended 40 embodiment of the invention, nanoparticles (such as albumin over an extended period of time, such as from about a month nanoparticles) of the inventive compounds can be adminis up to about seven years. In some embodiments, the compo tered by any acceptable route including, but not limited to, sition is administered over a period of at least about any of 2. orally, intramuscularly, transdermally, intravenously, through 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30, 36, 48, 60, 72, or 84 an inhaler or other airborne delivery systems and the like. In months. In some embodiments, the rapamycin or derivative 45 Some embodiments, the rapamycin orderivative thereof is not thereof is administered over a period of at least one month, coating a stent or is not administered using a stent. wherein the interval between each administration is no more The dosing frequency of the rapamycin-containing nano than about a week, and wherein the dose of the rapamycin or particle composition and the second compound may be a derivative thereof at each administration is about 0.25 adjusted over the course of the treatment based on the judg mg/m to about 75 mg/m, such as about 0.25 mg/m to about 50 ment of the administering physician. In some embodiments, 25 mg/m or about 25 mg/m to about 50 mg/m. the first and second therapies are administered simulta In some embodiments, the dosage of rapamycin in a nano neously, sequentially, or concurrently. When administered particle composition can be in the range of 100-400 mg/m separately, the rapamycin-containing nanoparticle composi when given on a 3 week schedule, or 50-250 mg/m when tion and the second compound can be administered at differ given on a weekly schedule. Preferably, the amount of rapa 55 ent dosing frequency or intervals. For example, the rapamy mycin is about 80 to about 180 mg/m (e.g., about 100 mg/m cin-containing nanoparticle composition can be administered to about 150 mg/m, such as about 120 mg/m). weekly, while a second compound can be administered more Other exemplary dosing schedules for the administration or less frequently. In some embodiments, Sustained continu of the nanoparticle composition (e.g., rapamycin/albumin ous release formulation of the rapamycin-containing nano nanoparticle composition) include, but are not limited to, 100 60 particle and/or second compound may be used. Various for mg/m, weekly, without break; 75 mg/m weekly, 3 out of mulations and devices for achieving Sustained release are four weeks; 100 mg/m, weekly, 3 out of4 weeks; 125 mg/m. known in the art. A combination of the administration con weekly, 3 out of 4 weeks; 125 mg/m, weekly, 2 out of 3 figurations described herein can be used. weeks; 130 mg/m, weekly, withoutbreak; 175 mg/m, once Modes of Administration of Combination Therapies every 2 weeks; 260 mg/m, once every 2 weeks; 260 mg/m. 65 In some embodiments, the present invention provides a once every 3 weeks; 180-300 mg/m, every three weeks; method of treating cancer comprising a first therapy compris 60-175 mg/m, weekly, withoutbreak; 20-150 mg/m twice a ing administering nanoparticles comprising rapamycin or a US 8,911,786 B2 45 46 derivative thereof and a carrier protein (e.g., albumin) and a and/or hormone therapeutic agent are administered with a second therapy comprising chemotherapy and/or hormone time separation of more than about 15 minutes, such as more therapy. In some embodiments, the method comprises a) a than about any of 20, 30, 40, 50, 60 or more minutes. Either first therapy comprising administering to the individual a the rapamycin or a derivative thereof nanoparticle composi composition comprising nanoparticles of rapamycin and an 5 tion or the chemotherapeutic agent and/or hormonetherapeu albumin; and b) a second therapy comprising chemotherapy tic agent may be administered first. The rapamycin or a and/or hormone therapy. derivative thereof nanoparticle composition and the chemo The dose of the inventive composition administered to an therapeutic agent and/or hormone therapeutic agent are con individual (e.g., a human) in combination therapy may vary tained in separate compositions, which may be contained in with the particular composition, the method of administra 10 the same or different packages. tion, and the particular stage of cancer being treated. The In Some embodiments, the administration of the rapamycin amount should be sufficient to produce a desirable response, or a derivative thereof nanoparticle composition and the che Such as a therapeutic or prophylactic response against cancer. motherapeutic agent and/or hormone therapeutic agent are In some embodiments, the amount of the composition is a concurrent, e.g., the administration period of the nanoparticle therapeutically effective amount. In some embodiments, that 15 composition and that of the chemotherapeutic agent and/or amount of the composition is a prophylactically effective hormone therapeutic agent overlap with each other. In some amount. In some embodiments, the amount of rapamycin or a embodiments, the administration of the rapamycin or a derivative thereof in the composition is below the level that derivative thereof nanoparticle composition and the chemo induces a toxicological effect (e.g., an effect above a clini therapeutic agent and/or hormone therapeutic agent are non cally acceptable level of toxicity) or is at a level where a concurrent. For example, in Some embodiments, the admin potential side effect can be controlled or tolerated when the istration of the rapamycin or a derivative thereof nanoparticle composition is administered to the individual. composition is terminated before the chemotherapeutic agent The composition comprising nanoparticles comprising and/or hormone therapy is administered. In some embodi rapamycin or a derivative thereof and a carrier protein (e.g., ments, the administration of the chemotherapeutic agent and/ albumin) (also referred to as "nanoparticle composition') and 25 or hormone therapy is terminated before the rapamycin or a the chemotherapeutic agent and/or hormone therapeutic derivative thereof nanoparticle composition is administered. agent can be administered simultaneously (e.g., simultaneous The time period between these two non-concurrent adminis administration) and/or sequentially (e.g., sequential admin trations can range from about two to eight weeks, such as istration). about four weeks. In some embodiments, the nanoparticle composition and 30 The dosing frequency of the rapamycin or a derivative the chemotherapeutic agent and/or hormone therapeutic thereof-containing nanoparticle composition and the chemo agents (including the specific chemotherapeutic agents therapeutic agent and/or hormone therapy may be adjusted described herein) are administered simultaneously. The term over the course of the treatment, based on the judgment of the 'simultaneous administration, as used herein, means that the administering physician. When administered separately, the nanoparticle composition and the chemotherapeutic agent 35 rapamycin or a derivative thereof-containing nanoparticle and/or hormone therapeutic agent are administered with a composition and the chemotherapeutic agent and/or hormone time separation of no more than about 15 minute(s). Such as therapy can be administered at different dosing frequency or no more than about any of 10, 5, or 1 minutes. When the drugs intervals. For example, the rapamycin or a derivative thereof. are administered simultaneously, the rapamycin or a deriva containing nanoparticle composition can be administered tive thereof in the nanoparticles and the chemotherapeutic 40 weekly, while a chemotherapeutic agent and/or hormone agent and/or hormonetherapeutic agent may be contained in therapeutic agent can be administered more or less frequently. the same composition (e.g., a composition comprising both In some embodiments, Sustained continuous release formu the nanoparticles and the chemotherapeutic agent) or in sepa lation of the rapamycin or a derivative thereof-containing rate compositions (e.g., the nanoparticles are contained in one nanoparticle and/or chemotherapeutic agent and/or hormone composition and the chemotherapeutic agent is contained in 45 therapeutic agent may be used. Various formulations and another composition). For example, rapamycin or a derivative devices for achieving Sustained release are known in the art. thereof and a carrier protein (e.g., albumin) and the chemo The rapamycin or a derivative thereof nanoparticle com therapeutic agent may be present in a single composition position and the chemotherapeutic agent and/or hormone containing at least two different nanoparticles, wherein some therapeutic agent can be administered using the same route of of the nanoparticles in the composition comprise rapamycin 50 administration or different routes of administration. In some or a derivative thereof and a carrier protein, and some of the embodiments (for both simultaneous and sequential admin other nanoparticles in the composition comprise the chemo istrations), the rapamycin or a derivative thereof in the nano therapeutic agent and a carrier protein. The invention contem particle composition and the chemotherapeutic agent and/or plates and encompasses such compositions. In some embodi hormone therapeutic agent are administered at a predeter ments, only rapamycin or a derivative thereof is contained in 55 mined ratio. For example, in Some embodiments, the ratio by nanoparticles. In some embodiments, simultaneous adminis weight of the rapamycin or a derivative thereof in the nano tration of the rapamycin or a derivative thereof in the nano particle composition and the chemotherapeutic agent or the particle composition and the chemotherapeutic agent and/or hormone therapeutic agent is about 1 to 1. In some embodi hormonetherapeutic agent can be combined with Supplemen ments, the weight ratio may be between about 0.001 to about tal doses of the rapamycin or a derivative thereof and/or the 60 1 and about 1000 to about 1, or between about 0.01 to about chemotherapeutic agent and/or hormone therapeutic agent. 1 and 100 to about 1. In some embodiments, the ratio by In some embodiments, the rapamycin or a derivative weight of the rapamycin or a derivative thereof in the nano thereof nanoparticle composition and the chemotherapeutic particle composition and the chemotherapeutic agent or hor agent and/or hormone therapeutic agent are administered mone therapeutic agent is less than any of about 100:1, 50:1, sequentially. The term "sequential administration' as used 65 30:1, 10:1, 9:1, 8:1, 7.5:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, and 1:1. herein means that the rapamycin or a derivative thereof in the In some embodiments, the ratio by weight of the rapamycin or nanoparticle composition and the chemotherapeutic agent a derivative thereof in the nanoparticle composition and the US 8,911,786 B2 47 48 chemotherapeutic agent or hormone therapeutic agent is The dose of the rapamycin or its derivative therein in the more than any of about 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 7.5:1, nanoparticle composition will vary with the nature of the 8:1, 9:1, 30:1, 50:1, 100:1. Other ratios are contemplated. combination therapy and the particular disease being treated. The doses required for the rapamycin or a derivative In some embodiments, the amount of rapamycin or a deriva thereof and/or the chemotherapeutic agent and/or hormone 5 tive thereof in the nanoparticle composition in the combina therapeutic agent may (but not necessarily) be lower than tion therapy is included in any of the following ranges: about what is normally required when each agent is administered 0.5 to about 5 mg, about 5 to about 10 mg, about 10 to about alone. Thus, in some embodiments, a Subtherapeutic amount 15 mg, about 15 to about 20 mg, about 20 to about 25 mg, of the rapamycin or a derivative thereof in the nanoparticle about 20 to about 50 mg, about 25 to about 50 mg, about 50 to composition and/or the chemotherapeutic agent and/or hor 10 mone therapeutic agent are administered. "Subtherapeutic about 75 mg, about 50 to about 100 mg, about 75 to about 100 amount’ or “subtherapeutic level” refer to an amount that is mg, about 100 to about 125 mg, about 125 to about 150 mg. less than the therapeutic amount, that is, less than the amount about 150 to about 175 mg, about 175 to about 200 mg, about normally used when the rapamycin or a derivative thereof in 200 to about 225 mg, about 225 to about 250 mg, about 250 to the nanoparticle composition and/or the chemotherapeutic 15 about 300 mg, about 300 to about 350 mg, about 350 to about agent and/or hormone therapeutic agent are administered 400 mg, about 400 to about 450 mg. or about 450 to about 500 alone. The reduction may be reflected in terms of the amount mg. In some embodiments, the amount of rapamycin or administered at a given administration and/or the amount derivative thereof in the effective amount of the nanoparticle administered overagiven period of time (reduced frequency). composition (e.g., a unit dosage form) for combination In some embodiments, enough chemotherapeutic agent therapy is in the range of about 54 mg to about 540 mg. Such and/or hormone therapeutic agent is administered so as to as about 180 mg to about 270 mg or about 216 mg. In some allow reduction of the normal dose of the rapamycin or a embodiments, the concentration of the rapamycin in the derivative thereof in the nanoparticle composition required to nanoparticle composition for use in combination therapy is effect the same degree of treatment by at least about any of dilute (about 0.1 mg/ml) or concentrated (about 100 mg/ml), 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more. In 25 including for example any of about 0.1 to about 50 mg/ml, Some embodiments, enough rapamycin ora derivative thereof about 0.1 to about 20 mg/ml, about 1 to about 10 mg/ml, about in the nanoparticle composition is administered so as to allow 2 mg/ml to about 8 mg/ml, about 4 to about 6 mg/ml, about 5 reduction of the normal dose of the chemotherapeutic agent mg/ml. In some embodiments, the concentration of the rapa and/or hormonetherapeutic agent required to affect the same mycin or a derivative thereof in the nanoparticle composition degree of treatment by at least about any of 5%, 10%, 20%, 30 in combination therapy is at least about any of 0.5 mg/ml, 1.3 30%, 50%, 60%, 70%, 80%, 90%, or more. mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/ml. 4 mg/ml, 5 mg/ml, 6 In some embodiments, the dose of both the rapamycin or a mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 derivative thereof in the nanoparticle composition and the mg/ml, 25 mg/ml, 30 mg/ml. 40 mg/ml, or 50 mg/ml. chemotherapeutic agent and/or hormone therapeutic agent Exemplary effective amounts of rapamycin or a derivative are reduced as compared to the corresponding normal dose of 35 thereof in the nanoparticle composition for use in combina each when administered alone. In some embodiments, both tion therapy include, but are not limited to, about any of 25 the rapamycin or a derivative thereof in the nanoparticle com mg/m, 30 mg/m, 50 mg/m, 60 mg/m, 75 mg/m, 80 position and the chemotherapeutic agent and/or hormone mg/m, 90 mg/m, 100 mg/m, 120 mg/m, 160 mg/m, 175 therapeutic agent are administered at a subtherapeutic, e.g., mg/m, 180 mg/m,200 mg/m, 210 mg/m, 220 mg/m,250 reduced level. In some embodiments, the dose of the nano 40 mg/m, 260 mg/m,300 mg/m,350 mg/m, 400 mg/m,500 particle composition and/or the chemotherapeutic agent is mg/m, 540 mg/m, 750 mg/m, 1000 mg/m, or 1080 mg/m Substantially less than the established maximum toxic dose rapamycin. In various embodiments, the rapamycin or a (MTD). For example, the dose of the rapamycin or a deriva derivative thereof in the nanoparticle composition includes tive thereof nanoparticle composition and/or the chemothera less than about any of 350 mg/m, 300 mg/m, 250 mg/m. peutic agent and/or hormone therapeutic agent is less than 45 200 mg/m, 150 mg/m, 120 mg/m, 100 mg/m, 90 mg/m. about 50%, 40%, 30%, 20%, or 10% of the MTD. 50 mg/m, or 30 mg/mrapamycin or a derivative thereof. In A combination of the administration configurations Some embodiments, the amount of the rapamycin or a deriva described herein can be used. The combination therapy meth tive thereof per administration in combination therapy is less ods described herein may be performed alone or in conjunc than about any of 25 mg/m, 22 mg/m. 20 mg/m, 18 mg/m. tion with another therapy, such as Surgery, radiation, gene 50 15 mg/m, 14 mg/m, 13 mg/m, 12 mg/m, 11 mg/m, 10 therapy, immunotherapy, bone marrow transplantation, stem mg/m, 9 mg/m, 8 mg/m, 7 mg/m, 6 mg/m, 5 mg/m, 4 cell transplantation, hormonetherapy, targeted therapy, cryo mg/m, 3 mg/m, 2 mg/m, or 1 mg/m. In some embodi therapy, ultrasound therapy, photodynamic therapy, and/or ments, the effective amount of rapamycin or a derivative chemotherapy and the like. Additionally, a person having a thereof in the composition for use in combination therapy is greater risk of developing the proliferative disease may 55 included in any of the following ranges: about 1 to about 5 receive treatments to inhibit or and/or delay the development mg/m, about 5 to about 10 mg/m, about 10 to about 25 of the disease. mg/m, about 25 to about 50 mg/m, about 50 to about 75 As will be understood by those of ordinary skill in the art, mg/m, about 75 to about 100 mg/m, about 100 to about 125 the appropriate doses of chemotherapeutic agents and/or hor mg/m, about 125 to about 150 mg/m, about 150 to about mone therapeutic agent will be approximately those already 60 175 mg/m, about 175 to about 200 mg/m, about 200 to employed in clinical therapies wherein the chemotherapeutic about 225 mg/m, about 225 to about 250 mg/m, about 250 agent and/or hormone therapeutic agent are administered to about 300 mg/m, about 300 to about 350 mg/m, or about alone or in combination with other chemotherapeutic agents. 350 to about 400 mg/m. Preferably, the effective amount of Variation in dosage will likely occur depending on the con rapamycin or a derivative thereof in the composition for use in dition being treated. As described above, in some embodi 65 combination therapy is about 30 to about 300 mg/m, such as ments, the chemotherapeutic agents and/or hormone thera about 100 to about 150 mg/m, about 120 mg/m, about 130 peutic agents may be administered at a reduced level. mg/m, or about 140 mg/m. US 8,911,786 B2 49 50 In some embodiments of any of the above aspects, the but are not limited to, 100 mg/m, weekly, withoutbreak; 75 effective amount of rapamycin or a derivative thereof in the mg/m weekly, 3 out of four weeks; 100 mg/m, weekly, 3 out nanoparticle composition for use in combination therapy of4 weeks; 125 mg/m, weekly, 3 out of 4 weeks; 125 mg/m. includes at least about any of 1 mg/kg, 2.5 mg/kg, 5 mg/kg, weekly, 2 out of 3 weeks; 130 mg/m, weekly, withoutbreak; 7.5 mg/kg, 10 mg/kg, 15 mg/kg, or 20 mg/kg. In various 175 mg/m, once every 2 weeks; 260 mg/m, once every 2 embodiments, the effective amount of rapamycin or a deriva weeks; 260 mg/m, once every 3 weeks; 180-300 mg/m. tive thereof in the nanoparticle composition for use in com every three weeks; 60-175 mg/m, weekly, without break; bination therapy includes less than about any of 350 mg/kg, 20-150 mg/m twice a week; and 150-250 mg/m twice a 300 mg/kg, 250 mg/kg, 200 mg/kg, 150 mg/kg, 100 mg/kg, week. The dosing frequency of the composition may be 50 mg/kg, 25 mg/kg, 20 mg/kg, 10 mg/kg, 5 mg/kg, or 1 10 adjusted over the course of the treatment based on the judg mg/kg rapamycin or a derivative thereof. ment of the administering physician. Exemplary dosing frequencies of rapamycin or a derivative The rapamycin or a derivative thereof nanoparticle com thereof in the nanoparticle composition for use in combina positions described herein can be administered to an indi tion therapy include, but are not limited to, weekly without vidual (such as human) during combination therapy via vari break; weekly, three out of four weeks; once every three 15 weeks; once every two weeks; weekly, two out of three ous routes, such as parenterally, including intravenous, intra weeks. In some embodiments, the rapamycin or a derivative arterial, intraperitoneal, intrapulmonary, oral, inhalation, thereof in the nanoparticle composition is administered in intravesicular, intramuscular, intra-tracheal, Subcutaneous, combination about once every 2 weeks, once every 3 weeks, intraocular, intrathecal, or transdermal. For example, the once every 4 weeks, once every 6 weeks, or once every 8 nanoparticle composition can be administered by inhalation weeks. In some embodiments, the rapamycin or a derivative to treat conditions of the respiratory tract. The rapamycin or a thereof in the nanoparticle composition is administered in derivative thereof nanoparticle compositions can be used to combination therapy at least about any of 1x, 2x. 3x, 4x, 5x, treat respiratory conditions such as pulmonary fibrosis, 6x, or 7x (i.e., daily) a week. In some embodiments, the broncheolitis obliterans, lung cancer, bronchoalveolar carci intervals between each administration in combination noma, and the like. In some embodiments, the nanoparticle therapy are less than about any of 6 months, 3 months, 1 25 compositions is administrated intravenously. In some month, 20 days, 15, days, 12 days, 10 days, 9 days, 8 days, 7 embodiments, the nanoparticle compositions is administered days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. In some orally. embodiments, the intervals between each administration in In some embodiments, the nanoparticle composition of the combination therapy are more than about any of 1 month, 2 rapamycin or a derivative thereof and the chemotherapeutic 30 agent is administered according to any of the dosing regimes months, 3 months, 4 months, 5 months, 6 months, 8 months, described in Table 1. or 12 months. In some embodiments, there is no break in the In some embodiments, there is provided a method of treat dosing schedule. In some embodiments, the interval between ing cancer in an individual comprising administering to the each administration is no more than about a week. individual: a) an effective amount of a composition compris The administration of the rapamycin or a derivative thereof ing nanoparticles comprising a rapamycin or a derivative in the nanoparticle composition in combination therapy can 35 thereof and an albumin, and b) an effective amount of at least be extended over an extended period of time, such as from one other chemotherapeutic agent as provided in Rows 1 to 53 about a month up to about seven years. In some embodiments, in Table 1. In some embodiments, the administration of the the rapamycin or a derivative thereof in the nanoparticle com nanoparticle composition and the chemotherapeutic agent position is administered over a period of at least about any of may be any of the dosing regimes as indicated in Rows 1 to 53 2,3,4,5,6,7,8,9, 10, 11, 12, 18, 24, 30, 36,48, 60, 72, or 84 40 in Table 1. In some embodiments, the cancer is early stage months. In some embodiments, the rapamycin or derivative cancer, non-metastatic cancer, primary cancer, advanced can thereof nanoparticle composition is administered over a cer, locally advanced cancer, metastatic cancer, cancer in period of at least one month, wherein the interval between remission, recurrent cancer, cancer in an adjuvant setting, each administration is no more than about a week, and cancerina neoadjuvant setting, or cancer Substantially refrac wherein the dose of the rapamycin or a derivative thereof in 45 tory to hormonetherapy. In some embodiments, the cancer is the nanoparticle composition at each administration is about a solid tumor. In some embodiments, the cancer is not a solid 0.25 mg/m to about 75 mg/m, such as about 0.25 mg/m to tumor (i.e., other than a solid tumor). In some embodiments, the cancer is a plasmacytoma. In some embodiments, the about 25 mg/m or about 25 mg/m to about 50 mg/m. cancer is multiple myeloma, renal cell carcinoma, prostate In some embodiments, the dosage of rapamycin in the cancer, lung cancer, melanoma, brain cancer (e.g., glioblas nanoparticle composition in the combination therapy can be 50 toma), ovarian cancer, or breast cancer. In some embodi in the range of 100-400 mg/m when given on a 3 week ments, the cancer is a carcinoma (i.e., other than a carci schedule, or 50-250mg/m when given on a weekly schedule. noma). In some embodiments, the cancer is not colon cancer Preferably, the amount of rapamycin is about 80 to about 180 (i.e., other than colon cancer). In some embodiments, the mg/m (e.g., about 100 mg/m to about 150 mg/m, such as cancer is not breast cancer (i.e., other than breast cancer). In about 120 mg/m). 55 Some embodiments, the cancer is not ovarian cancer, prostate Other exemplary dosing schedules for the administration cancer, or brain cancer. In some embodiments, one or more of the nanoparticle composition (e.g., rapamycin/albumin symptoms of the cancer are ameliorated. In some embodi nanoparticle composition) in combination therapy include, ments, the cancer is delayed or prevented. TABLE 1

Row No. Combination Regime. Dosage 1. RAPA + Carboplatin + RAPA: Rapamycin or a derivative thereof: any doses or Herceptin (R) regimes described above for combination therapy Carbo: AUC = 2 D1, 8, 15 q4wkx 6 Herceptin (R): 4 mg/kg on wk 1, 2 mg/kg all Subsequent weeks US 8,911,786 B2 51 52 TABLE 1-continued

Row No. Combination Regime. Dosage 2. RAPA alone RAPA: Rapamycin or a derivative thereof: any doses or (+Herceptin (R) regimes described above for combination therapy 3. RAPA + Navelbine (R) L1: Rapamycin or a derivative thereof: any doses or (G-CSF) regimes described above for combination therapy Nav: 15 mg/m. L2: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Nav: 20 mg/m’ L3: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy L4: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Nav: 25 mg/m. L5: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Nav: 25 mg/m. qwk all levels PA + Xeloda (R) RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Xeloda (R): 825 mg/m. D 1-14 q3wk RA PA + Anthracycline RA PA + RAPA: Rapamycin or a derivative thereof: any doses or Gemcitabine regimes described above for combination therapy Gem: 1000 mg/m2 qwk x 2/3 RA PA + Lapatinib Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Lapatinib: starting at 1000 mg/dx 2 days RA PA - FEC RAPA: Rapamycin or a derivative thereof: any doses or (+Herceptin (R) regimes described above for combination therapy FEC: 4 cycles (+Herceptin (R) for HER2+ pts) RA PA + Carboplatin + RAPA: Rapamycin or a derivative thereof: any doses or Avastin (R) regimes described above for combination therapy Carbo: AUC = 2 qwk D1, 8, 15 Avastin (R): 10 mg/m q2wk PA + Avastin (R) RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy + PA + Xeloda (R) + RAPA: Rapamycin or a derivative thereof: any doses or atinib regimes described above for combination therapy PA + Gemcitabine RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Gem: 1250 mg/m. D 1, 8 q3wk 3 PA - Sutent (R) RA PA + AC+ G AC+ G-CSF q2wkx 4 followed by RAPA: CSF (+Herceptin (R) Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy (+Herceptin (R) for HER2+ pts) RA PA + AC+ G Dose dense AC+ G-CSF followed by RAPA: CSF (+Herceptin (R) Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy (+Herceptin (R) for HER2+ pts) RA PA - AC AC followed by RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy RA PA - AC AC q2wk followed by RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy RX length 16 wks RA PA - AC Dose dense AC followed by RAPA: (+Avastin (R) Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy (+Avastin (R) in HER2+ pts) PA - AC AC (such as about 60 mg/m adriamycin and 600 mg/m’ cyclophosphamide, once every two weeks) followed by RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy 20. RA PA + AC + AC followed by RAPA: Rapamycin or a derivative thereof: Neulasta (R) any doses or regimes described above for combination therapy 21. PA - FEC RAPA: Rapamycin or a derivative thereof: any doses or erceptin (R) regimes described above for combination therapy followed by 5-FU: 500 mg/m (3wk Epirubicin: 100 mg/m’ (without Herceptin (R)) or Epirubicin: 75 mg/m. (with Herceptin (R) for HER2+ pts) Cyclophosphamide: 500 mg/m q3wk US 8,911,786 B2 53 54 TABLE 1-continued

Row No. Combination Regime. Dosage 22. RAPA Arm 1: Neoadjuvant: Gem: 2000 mg/m, RAPA: Rapamycin or Gemcitabine + a derivative thereof: any oses or regimes described above for Epirubicin combination therapy, Epi 50 mg/m q2wkx 6 Arm 2: Adjuvant: Gem: 2000 mg/m, RAPA: Rapamycin or a derivative thereof: any oses or regimes described above for combination therapy 23. RAPA + Herceptin (R) + RAPA: Rapamycin or a derivative thereof: any doses or Navelbine regimes described above for combination therapy + Herceptin (R) followed by Navelbine (R) + Herceptin (R) 24. RAPA + Carboplatin TAC vs AC followed by RAPA: (+Herceptin (R)) + AC Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy + carbo vs AC followed by RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination herapy + carbo + Herceptin (R) 25. RAPA + RAPA: Rapamycin or a derivative thereof: any doses or Capecitabine regimes described above for combination therapy Xeloda (R 850 mg/m. D 1-14 q3wkx 4 26. RAPA + Carboplatin RAPA: Rapamycin or a derivative thereof: any doses or (+Avastin (R) regimes described above for combination therapy Carbo qwk + Avastin (R) in HER2+ pts 27. RAPA + Carboplatin + RAPA: Rapamycin or a derivative thereof: any doses or Herceptin (R) + Avastin (R) regimes described above for combination therapy Carbo: AUC = 5 + Herceptin (R) + Avastin (R) 4 week cycle x 6 28. RAPA + Lapatinib RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Lapatinib: 1000 mg day 29. RAPA + RAPA: Rapamycin or a derivative thereof: any doses or Capecitabine regimes described above for combination therapy Xeloda (R): 1000 mg/m D 1-14 q3wkx 4 30. RAPA+Avastin (R) + RAPA: Rapamycin or a derivative thereof: any doses or AC (+G-CSF) regimes described above for combination therapy it Avastin (R) ollowed by Aqwk + C daily 31. RAPA-AC RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy followed 32. RAPA + Carboplatin + RAPA: Rapamycin or a derivative thereof: any doses or Avastin (R) regimes described above for combination therapy Carbo: AUC = 6 a.3wk Avastin (R): 15 mg/kg 33. RA PA + Carboplatin RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Carbo fixed at AUC = 6 awk 34. RA PA + Carboplatin + Avastin (R) 35. RA PA + Gemcitabine or PA + Avastin (R) 36. PA + Carboplatin + RAPA: Rapamycin or a derivative thereof: any doses or Avastin (R) regimes described above for combination therapy Carbo: AUC = 6 q3 wk+ Avastin (R) 37. RAPA + Alimta (R) RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy : 500 mg q3wk 38. PA + Cisplatin 39. PA + Navelbine (R) + Cisplatin 40. RAPA + Carboplatin RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Carbo: AUC = 6 q3wk 41. RAPA + Carboplatin RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Carbo: AUC = 6 42. RAPA + Avastin (R) RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Avastin (R): 10 mg/m q2wk 43. RAPA - 5-FU RAPA: Rapamycin or a derivative thereof: any doses or Cisplatin regimes described above for combination therapy 5-FU:750 mg/m2 CIVx 5 cisplatin: 75 mg/m. D 1 ollowed by XRT surgery RAPA + Cetuximab 45. RAPA + 46. RAPA RAPA: Rapamycin or a derivative thereof: any doses or Gemcitabine regimes described above for combination therapy Gemcitabine: 1000 mg/m. D 1 and D 8 US 8,911,786 B2 55 56 TABLE 1-continued Row No. Combination Regime. Dosage 47. RAPA + Gefitinib RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination therapy Gefitinib starting at 1000 mg/dx 2 48. RAPA + Sorafenib + RAPA: Rapamycin or a derivative thereof: any doses or Carboplatin regimes described above for combination therapy Sorafenib: D2-19 Carbo: AUC = 6 D1 49. RAPA + RAPA: Rapamycin or a derivative thereof: any doses or Capecitabine regimes described above for combination therapy + Xeloda (R) at a range of about 500-2500 mg/m (such as any of about 550 mg/m, 650 mg/m, 85 mg/m, 850 mg/m, 100 mg/m, 1250 mg/m) SO. RAPA + Weekly Gemcitabine S1. RAPA+ anti angiogenic agent(s) 52. RAPA + proteasome inhibitor(s) 53. RAPA + tyrosine kinase inhibitor(s) 54. RAPA - EGFR inhibitor(s)

As used in herein (for example in Table 1), RAPA refers to rapamycin or a derivative thereof nanoparticles and the radia a composition comprising nanoparticles comprising rapamy 25 tion/surgery would still be able to exert an advantageously cin or a derivative thereofanda carrier protein (e.g., albumin); combined effect on the cell. For example, the rapamycin or GW572016 refers to lapatinib: Xel refers to capecitabine or derivative thereof in the nanoparticle composition may be Xeloda(R); bevacizumab is also known as Avastin R; trastu administered less than about any of 1, 3, 6, 9, 12, 18, 24, 48, Zumab is also known as Herceptin R; pemetrexed is also 30 60, 72, 84, 96, 108, 120 hours prior to the radiation and/or known as Alimta R, cetuximab is also known as Erbitux(R); Surgery. In some embodiments, the nanoparticle composition gefitinib is also known as Iressa(R); FEC refers to a combina is administered less than about 9 hours prior to the radiation tion of 5-fluorouracil, Epirubicin and Cyclophosphamide: AC and/surgery. In some embodiments, the nanoparticle compo refers to a combination of Adriamycin plus Cyclophospha sition is administered less than about any of 1, 2, 3, 4, 5, 6, 7, mide. 35 8, 9, or 10 days prior to the radiation/surgery. In some As used herein (for example in Table 1), AUC refers to area embodiments, the rapamycin or a derivative thereof in the under curve; q4wk refers to a dose every 4 weeks; q3wk refers nanoparticle composition is administered less than about any to a dose every 3 weeks; q2wk refers to a dose every 2 weeks: of 1, 3, 6, 9, 12, 18, 24, 48, 60, 72, 84, 96, 108, or 120 hours qwk refers to a weekly dose; qwkx 3/4 refers to a weekly dose after the radiation and/or Surgery. In some embodiments, it for 3 weeks with the 4" week off;qwkx 2/3 refers to a weekly 40 may be desirable to extend the time period for treatment dose for 2 weeks with the 3" week off. significantly, where several days to several weeks lapse In some embodiments, the present invention provides a between the two therapies. method of treating cancer comprising a first therapy compris Radiation contemplated herein includes, for example, ing administering nanoparticles comprising rapamycin or a Y-rays, X-rays (external beam), and the directed delivery of derivative thereof and a carrier protein (e.g., albumin) and a 45 radioisotopes to tumor cells. Other forms of DNA damaging second therapy comprising Surgery, radiation, gene therapy, factors are also contemplated Such as microwaves and UV immunotherapy, bone marrow transplantation, stem cell irradiation are also contemplated. Radiation may be given in transplantation, targeted therapy, cryotherapy, ultrasound a single dose or in a series of Small doses in a dose-fraction therapy, and/or photodynamic therapy. In some embodi ated schedule. The amount of radiation contemplated herein ments, the method comprises a) a first therapy comprising 50 ranges from about 1 to about 100 Gy, including, for example, administering to the individual a composition comprising about 5 to about 80, about 10 to about 50 Gy, or about 10 Gy. nanoparticles of rapamycin and an albumin; and b) a second The total dose may be applied in a fractioned regime. For therapy comprising Surgery, radiation, gene therapy, immu example, the regime may comprise fractionated individual notherapy, bone marrow transplantation, stem cell transplan doses of 2 Gy. Dosage ranges for radioisotopes vary widely, tation, targeted therapy, cryotherapy, ultrasound therapy, and/ 55 and depends on the half-life of the isotope and the strength or photodynamic therapy. In some embodiments, the cancer and type of radiation emitted. may be prostate cancer. In some embodiments, the second When the radiation comprises use of radioactive isotopes, therapy is radiation therapy. In some embodiments, the sec the isotope may be conjugated to a targeting agent, such as a ond therapy is Surgery. therapeutic antibody, which carries the radionucleotide to the The administration of the rapamycin or a derivative thereof 60 target tissue. Suitable radioactive isotopes include, but are not nanoparticle composition may be prior to the radiation and/or limited to, astatine'', carbon'', chromium', chlorine, Surgery, after the radiation and/or Surgery, or concurrent with iron, cobalt, copper'7,67 Eu'', gallium', hydrogen, the radiation and/or Surgery. For example, the administration iodine'', iodine'', indium", iron, phosphorus, rhe of the nanoparticle composition may precede or follow the nium', selenium', sulphur, technicium'", and/or radiation and/or Surgery therapy by intervals ranging from 65 yttrium'. minutes to weeks. In some embodiments, the time period In some embodiments, enough radiation is applied to the between the first and the second therapy is such that the individualso as to allow reduction of the normal dose of the US 8,911,786 B2 57 58 rapamycin or a derivative thereof in the nanoparticle compo prising administering to the individual a composition com sition required to affect the same degree of treatment by at prising nanoparticles comprising rapamycin or a derivative least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, thereof and an albumin; and b) a second therapy comprising 80%, 90%, or more. In some embodiments, enough rapamy radiation as provided in Rows 1 to 11 in Table 2. In some cin or derivative thereof in the nanoparticle composition is 5 embodiments, the administration of the nanoparticle compo administered so as to allow reduction of the normal dose of sition and the chemotherapeutic agent may be any of the the radiation required to affect the same degree of treatment dosing regimes as indicated in Rows 1 to 11 in Table 2. In by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, Some embodiments, the cancer is early stage cancer, non 80%, 90%, or more. In some embodiments, the dose of both metastatic cancer, primary cancer, advanced cancer, locally the rapamycin or a derivative thereof in the nanoparticle com 10 advanced cancer, metastatic cancer, cancer in remission, position and the radiation are reduced as compared to the recurrent cancer, cancer in an adjuvant setting, cancer in a corresponding normal dose of each when used alone. neoadjuvant setting, or cancer Substantially refractory to hor In some embodiments, the combination of administration mone therapy. In some embodiments, the cancer is a solid of the rapamycin or a derivative thereof nanoparticle compo tumor. In some embodiments, the cancer is not a solid tumor sition and the radiation therapy produce Supra-additive effect. 15 (i.e., other than a Solid tumor). In some embodiments, the In some embodiments, the rapamycin or a derivative thereof cancer is a plasmacytoma. In some embodiments, the cancer in the nanoparticle composition is administered once at the is multiple myeloma, renal cell carcinoma, prostate cancer, dose of about 50 mg to 540 mg or about 30 mg/m to 300 mg/m, and the radiation is applied five times at 80 Gy daily. lung cancer, melanoma, brain cancer (e.g., glioblastoma). Administration of rapamycin or a derivative thereof nano 20 ovarian cancer, or breast cancer. In some embodiments, the particle compositions disclosed above in conjunction with cancer is a carcinoma (i.e., other than a carcinoma). In some administration of chemotherapeutic agent and/or hormone embodiments, the cancer is not colon cancer (i.e., other than therapeutic agentis equally applicable to those in conjunction colon cancer). In some embodiments, the cancer is not breast with radiation therapy and/or Surgery. cancer (i.e., other than breast cancer). In some embodiments, In some embodiments, the nanoparticle composition of the * the cancer is not ovarian cancer, prostate cancer, or brain rapamycin or a derivative thereof nanoparticles and/or the CaCC. TABLE 2

Row No. Combination Regime. Dosage 1 RAPA + Radiation RAPA + Carboplatin + Radiation RAPA + Carboplatin + cycle RAPA/Carbo induction followed by Radiation 2 or 3 times weekly pulse RAPA + radiation RAPA + Carboplatin + Radiation RAPA + Carboplatin + RAPA: Rapamycin or a derivative thereof: Radiation any doses or regimes described above for combination therapy + carbo + radiation ollowed by RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination herapy + carbo 6 RAPA + Radiation RAPA + Cetuximab + Radiation RAPA + Carboplatin + induction: RAPA: Rapamycin or a derivative 5-FU + Hydroxyurea + hereof: any doses or regimes described above Radiation or combination therapy + carbo: AUC = 2 followed by Concurrent chemoradiation: RAPA: Rapamycin or a derivative thereof: any doses or regimes described above for combination herapy: 5-FU: 600 mg/m; hydroxyurea: 5000 mg BID RAPA + Carboplatin + RAPA: Rapamycin or a derivative thereof: Erbitux (R) + Radiation any doses or regimes described above for combination therapy Eribitux (R): 400 mg/m·day 7, 250 mg/m’ qwkx 7 Carbo: AUC = 1.5 qwk x 7 MRT 10 RAPA + Gemcitabine + Radiation 11 RAPA + Cisplatin + Radiation chemotherapeutic agent is administered in conjunction with Metronomic Therapy Regimes radiation according to any of the dosing regimes described in The present invention also provides metronomic therapy Table 2. 65 regimes for any of the methods of treatment and methods of In some embodiments, there is provided a method of treat administration described herein. Exemplary metronomic ing cancer in an individual comprises a) a first therapy com therapy regimes and embodiments for the use of metronomic US 8,911,786 B2 59 60 therapy regimes are discussed below and disclosed in U.S. Carrier Proteins Ser. No. 1 1/359,286, filed Feb. 21, 2006, published as U.S. Provide herein are compositions comprising nanoparticles Pub. No. 2006/0263434 (such as those described in para that comprise rapamycin and a carrier protein for use methods graphs 0138 to 0157), which is hereby incorporated by of treatment of cancer, methods of administration, and dosage reference in its entirety. In some embodiments, the nanopar regimes described herein. In some embodiments, rapamycin ticle composition is administered over a period of at least one may be rapamycin or its derivatives or pharmaceutically month, wherein the interval between each administration is acceptable salts and accordingly the invention contemplates no more than about a week, and wherein the dose of rapamy and includes all these embodiments. In some embodiments, cin or a derivative thereof at each administration is about the carrier protein is albumin. In some embodiments, the 0.25% to about 25% of its maximum tolerated dose following 10 carrier protein is human serum albumin. a traditional dosing regime. In some embodiments, the nano Examples of Suitable carrier proteins include proteins nor particle composition is administered over a period of at least mally found in blood or plasma, which include, but are not two months, wherein the interval between each administra limited to, albumin, immunoglobulin including IgA, lipopro tion is no more than about a week, and wherein the dose of 15 teins, apolipoprotein B, C-acid glycoprotein, B-2-macroglo rapamycin or a derivative thereof at each administration is bulin, thyroglobulin, transferin, fibronectin, factor VII, factor about 1% to about 20% of its maximum tolerated dose fol VIII, factor IX, factor X, and the like. In some embodiments, lowing a traditional dosing regime. In some embodiments, the the carrier protein is a non-blood protein, such as casein, dose of rapamycin or a derivative thereofper administration is C.-lactalbumin, or B-lactoglobulin. The carrier proteins may less than about any of 25%, 24%, 23%, 22%, 20%, 18%, 15%, either be natural in origin or synthetically prepared. In some 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%. 5%, 4%, 3%, embodiments, the pharmaceutical acceptable carrier com 2%, or 1% of the maximum tolerated dose. In some embodi prises albumin, such as human serum albumin (HSA). HSA is ments, the nanoparticle composition is administered at least a highly soluble globular protein of M, 65K and consists of about any of 1x, 2x, 3x, 4x, 5x, 6.x, or 7x (i.e., daily) a week. 585 amino acids. HSA is the most abundant protein in the In some embodiments, the intervals between each adminis 25 plasma and accounts for 70-80% of the colloid osmotic pres tration are less than about any of 6 months, 3 months, 1 Sure of human plasma. The amino acid sequence of HSA month, 20 days, 15, days, 12 days, 10 days, 9 days, 8 days, 7 contains a total of 17 disulphide bridges, one free thiol (Cys days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. In some 34), and a single tryptophan (Trp 214). Other albumins are embodiments, the intervals between each administration are contemplated, such as bovine serum albumin. Use of Such 30 non-human albumins could be appropriate, for example, in more than about any of 1 month, 2 months, 3 months, 4 the context of use of these compositions in non-human mam months, 5 months, 6 months, 8 months, or 12 months. In some mals, such as the veterinary animals (including domestic pets embodiments, the composition is administered over a period and agricultural animals). of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, Human serum albumin (HSA) has multiple hydrophobic 30, 36, 48, 60, 72, or 84 months. 35 binding sites (a total of eight for fatty acids, an endogenous Pharmaceutical Agents ligand of HSA) and binds a diverse set of drugs, especially Provided herein are compositions comprising nanopar neutral and negatively charged hydrophobic compounds ticles that comprise rapamycin for use in the methods of (Goodman et al., The Pharmacological Basis of Therapeu treatment of cancer, methods of administration, and dosing tics, 9" ed. McGraw-Hill New York (1996)). Two high affin regimes described herein. In some embodiments, rapamycin 40 ity binding sites have been proposed in Subdomains IIA and may be rapamycin or its derivatives or pharmaceutically IIIA of HSA, which are highly elongated hydrophobic pock acceptable salts and accordingly the invention contemplates ets with charged lysine and arginine residues near the Surface and includes all these embodiments. Rapamycin is some which function as attachment points for polar ligand features times referred to elsewhere as Sirolimus, rapammune, or rapa (see, e.g., Fehske et al., Biochem. Pharmcol., 30, 687-92 mune. Derivatives of rapamycin include, but are not limited 45 (1981), Vorum, Dan. Med. Bull., 46, 379-99 (1999), Kragh to, compounds that are structurally similar to rapamycin or Hansen, Dan. Med. Bull., 1441, 131-40 (1990), Curry et al., are in the same general chemical class as rapamycin. Nat. Struct. Biol., 5, 827-35 (1998), Sugio et al., Protein. In some embodiments, the derivative of rapamycin retains Eng., 12, 439-46 (1999). He et al., Nature, 358, 209-15 one or more similar biological, pharmacological, chemical (1992), and Carter et al., Adv. Protein. Chem., 45, 153-203 and/or physical properties (including, for example, function 50 (1994)). ality) as rapamycin. In some embodiments, the rapamycin The carrier protein (e.g., albumin) in the composition gen derivative has at least about any of 10%, 20%, 30%, 40%, erally serves as a carrier for rapamycin or derivative thereof, 50%. 60%, 70%, 80%, 90%. 95% or 100% of an activity of i.e., the carrier protein in the composition makes the rapamy rapamycin. For example, the decrease in the size of a tumor, cin orderivative thereof more readily Suspendable in an aque the number of cancer cells, or the growth rate of a tumor 55 ous medium or helps maintain the Suspension as compared to caused by a rapamycin derivative is preferably at least about compositions not comprising a carrier protein. This can avoid any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, the use of toxic solvents for solubilizing rapamycin or a 95% or 100% of the corresponding decrease caused by the derivative thereof, and thereby can reduce one or more side same amount of rapamycin. An exemplary rapamycin deriva effects of administration of rapamycin or a derivative thereof tive includes benzoyl rapamycin, such as that disclosed in 60 into an individual (e.g., human). In some embodiments, the paragraph (0022 of WO 2006/089207, which is hereby composition is Substantially free (e.g. free) of organic Sol incorporated by reference in its entirety. Other exemplary vents or surfactants. A composition is “substantially free of rapamycin derivatives include WY-090217, AY-22989, NSC organic solvent' or “substantially free of surfactant if the 226080, SiiA-9268A, oxaazacyclohentriacontine, temsiroli amount of organic solvent or Surfactant in the composition is mus (CCI-779 (Wyeth)), everolimus (RAD001 (Novartis)), 65 not sufficient to cause one or more side effect(s) in an indi pimecrolimus (ASM981), SDZ-RAD, SAR943, ABT-578, vidual when the composition is administered to the indi AP23573, and Biolimus A9. vidual. US 8,911,786 B2 61 62 Rapamycin is “stabilized in an aqueous Suspension if it shape) with an average or mean diameter of no greater than remains suspended in an aqueous medium (e.g., without vis about 1000 nanometers (nm). Such as no greater than about ible precipitation or sedimentation) for an extended period of any of 900 nm, 800 nm, 700 nm, 600 nm, 500 nm, 400 nm, time, such as for at least about any of 0.1, 0.2,0.25, 0.5, 1, 2, 300 nm, 200 nm, or 100 nm. In some embodiments, the 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or 72 hours. The average or mean diameter of the particles is no greater than Suspension is generally, but not necessarily, Suitable for about 200 nm. In some embodiments, the average or mean administration to an individual (e.g., human). Stability of the diameter of the particles is between about 20 to about 400 nm. Suspension is generally (but not necessarily) evaluated at In some embodiments, the average or mean diameter of the storage temperature, such as room temperature (e.g., 20-25° particles is between about 40 to about 200 nm. In some C.) or refrigerated conditions (e.g., 4° C.). For example, a 10 embodiments, the particles are sterile-filterable. Suspension is stable at a storage temperature if it exhibits no In some embodiments, the nanoparticles comprise the flocculation or particle agglomeration visible to the naked eye rapamycin or a derivative thereof coated with a coating com or when viewed under the optical microscope at 1000 times, prising the carrier protein (such as albumin). In some embodi at about fifteen minutes after preparation of the Suspension. ments, the coating consists essentially of or consists of the Stability can also be evaluated under accelerated testing con 15 carrier protein. In some embodiments, at least a portion of the ditions, such as at a temperature that is higher than about 40° carrier protein in the nanoparticle portion of the rapamycin C. (or rapamycin derivative) nanoparticle composition is In some embodiments, the composition comprises nano crosslinked (for example crosslinked by disulfide bonds). particles comprising (in various embodiments consisting The nanoparticles described herein may be present in a dry essentially of) rapamycin and a carrier protein. When rapa formulation (e.g., lyophilized composition) or Suspended in a mycin is in a liquid form, the particles or nanoparticles are biocompatible medium. Suitable biocompatible media also referred to as droplets or nanodroplets. In some embodi include, but are not limited to, water, buffered aqueous media, ments, rapamycin is coated with the carrier protein. Particles saline, buffered saline, optionally buffered solutions of amino (such as nanoparticles) of poorly water Soluble pharmaceuti acids, optionally buffered solutions of proteins, optionally cal agents have been disclosed in, for example, U.S. Pat. Nos. 25 buffered solutions of sugars, optionally buffered solutions of 5,916,596; 6,506,405; and 6,537,579 and also in U.S. Pat. vitamins, optionally buffered solutions of synthetic poly App. Pub. No. 2005/0004002A1. mers, lipid-containing emulsions, and the like. The amount of carrier protein in the composition described In some embodiments, the nanoparticles do not comprise a herein will vary depending on the rapamycin or derivative blood-insoluble gas or do not comprise gas-filled thereof and other components in the composition. In some 30 microbubbles. embodiments, the composition comprises a carrier protein in The amount of carrier protein in the composition described an amount that is sufficient to stabilize the rapamycin in an herein will vary depending on the rapamycin or derivative aqueous Suspension, for example, in the form of a stable thereof and other components in the composition. In some colloidal Suspension (e.g., a stable Suspension of nanopar embodiments, the composition comprises a carrier protein in ticles). In some embodiments, the carrier protein is in an 35 an amount that is sufficient to stabilize the rapamycin in an amount that reduces the sedimentation rate of rapamycin in aqueous Suspension, for example, in the form of a stable an aqueous medium. For particle-containing compositions, colloidal Suspension (e.g., a stable Suspension of nanopar the amount of the carrier protein also depends on the size and ticles). In some embodiments, the carrier protein is in an density of particles of rapamycin. amount that reduces the sedimentation rate of rapamycin in In some embodiments of any of the aspects of the inven 40 an aqueous medium. The amount of the carrier protein also tion, the rapamycin or a derivative thereof is coated with a depends on the size and density of particles of rapamycin. carrier protein, Such as albumin (e.g., human serum albumin). Also provided herein are methods of reducing side effects In various embodiments, the composition comprises more associated with administration of a poorly water soluble phar than about any of 50%, 60%, 70%, 80%, 90%, 95%, or 99% maceutical agent to a human, comprising administering to a of the rapamycin orderivative thereof in nanoparticle form. In 45 human a pharmaceutical composition comprising the poorly Some embodiments, the rapamycin orderivative thereof con water soluble pharmaceutical agent, and a biocompatible stitutes more than about any of 50%, 60%, 70%, 80%, 90%, polymer (such as a carrier protein). For example, the inven 95%, or 99% of the nanoparticles by weight. In some embodi tion provides methods of reducing various side effects asso ments, the nanoparticle has a non-polymeric matrix. In some ciated with administration of the poorly water soluble phar embodiments, the rapamycin or derivative thereof is in an 50 maceutical agent, including, but not limited to, anhydrous, amorphous, and/or non-crystalline form. In some myelosuppression, neurotoxicity, hypersensitivity, inflam embodiments, the rapamycin or derivative thereof is amor mation, Venous irritation, phlebitis, pain, skin irritation, neu phous. In some embodiments, the nanoparticles comprise a tropenic fever, anaphylactic reaction, hematologic toxicity, core of rapamucin or derivative thereof that is substantially and cerebral or neurologic toxicity, and combinations thereof. free of polymeric materials (such as polymeric matrix). 55 In some embodiments, there is provided a method of reducing In some embodiments, the albumin to rapamycin weight hypersensitivity reactions associated with administration of ratio in the nanoparticles or in the nanoparticle composition is rapamycin or a derivative thereof, including, for example, about any of 18:1 or less, 15:1 or less, 14:1 or less, 13:1 or severe skin rashes, hives, flushing, dyspnea, tachycardia, can less, 12:1 or less, 11:1 or less, 10:1 or less, 9:1 or less, 8:1 or cer (e.g., lymphoma); chest pain; black, tarry stools; general less, 7.5:1 or less, 7:1 or less, 6:1 or less, 5:1 or less, 4:1 or 60 feeling of illness, shortness of breath; Swollen glands; weight less, or 3:1 or less. In some embodiments, the composition loss; yellow skin and eyes, abdominal pain; unexplained anx comprises a stable aqueous Suspension of particles (e.g., iousness; bloody or cloudy urine; bone pain; chills; confu nanoparticles) comprising rapamycin or a derivative thereof sion; convulsions (seizures); cough; decreased urge to uri and albumin (e.g., particles of rapamycin or a derivative nate; fast, slow, or irregular heartbeat; fever, frequent urge to thereof coated with albumin). 65 urinate; increased thirst; loss of appetite; lower back or side In some embodiments, the composition comprises nano pain; mood changes; muscle pain or cramps; nausea or Vom particles of any shape (e.g., a spherical or non-spherical iting; numbness ortingling around lips, hands, or feet; painful US 8,911,786 B2 63 64 or difficulturination; rash.; Sore throat; Sores or white spots on Sugar is not contained or used in the methods of treatment, lips or in mouth; Swelling of hands, ankles, feet, or lower legs; methods of administration, and dosage regimes described Swollen glands; trouble breathing; unusual bleeding or bruis herein ing; unusual tiredness or weakness; weakness or heaviness of Stabilizing Agents in Composition legs, skin ulcer or Sores, weight gain, acne; constipation; 5 In some embodiments, the compositions of the invention diarrhea, difficulty in moving; headache; loss of energy or also include a stabilizing agent for use in the methods of weakness; muscle pain or stiffness; pain; shaking or trem treatment, methods of administration, and dosage regimes bling; trouble sleeping; nosebleed; and/or Swelling of the described herein. In some embodiments, the compositions of face. These side effects, however, are merely exemplary and the invention include an antimicrobial agent and/or a Sugar 10 and/or a stabilizing agent for use in the methods of treatment, other side effects, or combination of side effects, associated methods of administration, and dosage regimes described with rapamycin can be reduced. The side effects may be herein. Exemplary stabilizing agents and embodiments for immediate or delayed (such as not occurring for a few days, the use of stabilizing agents are disclosed in U.S. Ser. No. weeks, months, or years after treatment begins). 11/513,756, filed Aug. 30, 2006 (such as those described in Antimicrobial Agents in Compositions 15 paragraphs 0038 to 0083) and 0107 to 0114). The In some embodiments, the compositions of the invention present invention in one of its embodiments provides for also includes an antimicrobial agent (e.g., an agent in addition compositions and methods of preparation of rapamycin to the rapamycin orderivative thereof in an amount Sufficient which retain the desirable therapeutic effects and remain to significantly inhibit (e.g., delay, reduce, slow, and/or pre physically and/or chemically stable upon exposure to certain Vent) microbial growth in the composition for use in the conditions such as prolonged storage, elevated temperature, methods of treatment, methods of administration, and dosage or dilution for parenteral administration. The stabilizing regimes described herein. Exemplary microbial agents and agent includes, for example, chelating agents (e.g., citrate, embodiments for the use of microbial agents are disclosed in malic acid, edetate, or pentetate), sodium pyrophosphate, and U.S. Ser. No. 11/514,030, filed Aug. 30, 2006 (such as those Sodium gluconate. In one embodiment, the invention pro described in paragraphs 0036 to 0058). In some embodi 25 vides pharmaceutical formulations of rapamycin or a deriva ments, the antimicrobial agent is a chelating agent, such as tive thereof comprising citrate, Sodium pyrophosphate, EDTA, edetate, citrate, pentetate, tromethamine, Sorbate, EDTA, Sodium gluconate, citrate and sodium chloride, and/or ascorbate, derivatives thereof, or mixtures thereof In some a derivative thereof. In another embodiment, the invention embodiments, the antimicrobial agent is a polydentate chelat provides a composition of rapamycin comprising a Surfac ing agent. In some embodiments, the antimicrobial agent is a 30 tant, wherein the rapamycin used for preparing the formula non-chelating agent, Such as any of Sulfites, benzoic acid, tion is in an anhydrous form prior to being incorporated into benzyl alcohol, chlorobutanol, paraben, orderivatives thereof the composition. In some embodiments, an antimicrobial other than rapamycin In some embodiments, a stabilizing agent is not contained orderivatives thereofdiscussed above is not contained or used or used in the methods of treatment, methods of administra in the methods of treatment, methods of administration, and 35 tion, and dosage regimes described herein. dosage regimes described herein Pharmaceutical Compositions and Formulations Sugar Containing Composition The compositions described herein may be used in the In some embodiments, the compositions of the invention preparation of a formulation, such as a pharmaceutical for include a Sugar for use in the methods of treatment described mulation, by combining the nanoparticle composition(s) herein. In some embodiments, the compositions of the inven 40 described with a pharmaceutical acceptable carrier, excipi tion include both a Sugar and an antimicrobial agent for use in ents, stabilizing agents or other agent, which are known in the the methods of treatment described herein. Exemplary sugars art, for use in the methods of treatment, methods of adminis and embodiments for the use of Sugars are disclosed in U.S. tration, and dosage regimes described herein. In some Ser. No. 11/514,030, filed Aug. 30, 2006 (such as those embodiments, the pharmaceutical composition includes described in paragraphs 0084 to 0090). In some embodi 45 nanoparticles comprising rapamycin or a derivative thereof ments, the Sugar serves as a reconstitution enhancer which and a carrier protein (e.g., albumin). In some embodiments, causes a lyophilized composition to dissolve or Suspend in the pharmaceutical composition includes a) nanoparticles water and/or aqueous solution more quickly than the lyo comprising rapamycin or a derivative thereof and a carrier philized composition would dissolve without the Sugar. In protein (e.g., albumin) and b) at least one other therapeutic Some embodiments, the composition is a liquid (e.g., aque 50 agent. In some embodiments, the other therapeutic agent ous) composition obtained by reconstituting or resuspending comprises a chemotherapeutic agent (Such as any of the che a dry composition. In some embodiments, the concentration motherapeutic agents described herein). In some embodi of Sugar in the composition is greater than about 50 mg/ml. In ments, the other therapeutic agent comprises a hormone Some embodiments, the Sugar is in an amount that is effective therapeutic agent. to increase the stability of the rapamycin orderivative thereof 55 To increase stability by increasing the negative Zeta poten in the composition as compared to a composition without the tial of nanoparticles, certain negatively charged components Sugar. In some embodiments, the Sugar is in an amount that is may be added. Such negatively charged components include, effective to improve filterability of the composition as com but are not limited to bile salts, bile acids, glycocholic acid, pared to a composition without the Sugar. cholic acid, chenodeoxycholic acid, taurocholic acid, glyco The Sugar-containing compositions described herein may 60 chenodeoxycholic acid, taurochenodeoxycholic acid, lito further comprise one or more antimicrobial agents, such as cholic acid, urSodeoxycholic acid, dehydrocholic acid, and the antimicrobial agents described herein or in U.S. Ser. No. others; phospholipids including lecithin (egg yolk) based 11/514,030, filed Aug. 30, 2006. In addition to one or more phospholipids which include the following phosphatidylcho Sugars, other reconstitution enhancers (such as those lines: palmitoyloleoylphosphatidylcholine, palmitoyllino described in U.S. Pat. App. Publication No. 2005/0152979, 65 leoylphosphatidylcholine, Stearoyllinoleoylphosphati which is hereby incorporated by reference in its entirety) can dylcholine, Stearoyloleoylphosphatidylcholine, also be added to the compositions. In some embodiments, a Stearoylarachidoylphosphatidylcholine, and dipalmi US 8,911,786 B2 65 66 toylphosphatidylcholine. Other phospholipids including example pH ranges of any of about 5.0 to about 8.0, about 6.5 L-O-dimyristoylphosphatidylcholine (DMPC), dio to about 7.5, and about 6.5 to about 7.0. In some embodi leoylphosphatidylcholine (DOPC), distearoylphosphatidyl ments, the pH of the composition is formulated to no less than choline (DSPC), hydrogenated soy phosphatidylcholine about 6, including for example no less than about any of 6.5, (HSPC), and other related compounds. Negatively charged 5 7, or 8 (e.g., about 8). The composition can also be made to be Surfactants or emulsifiers are also suitable as additives, e.g., isotonic with blood by the addition of a suitable tonicity sodium cholesteryl sulfate and the like. modifier, such as glycerol. In some embodiments, the composition is Suitable for The nanoparticles of this invention can be enclosed in a administration to a human. In some embodiments, the com hard or soft capsule, can be compressed into tablets, or can be position is suitable for administration to a mammal Such as, in 10 incorporated with beverages or food or otherwise incorpo the veterinary context, domestic pets and agricultural ani rated into the diet. Capsules can be formulated by mixing the mals. There are a wide variety of suitable formulations of the nanoparticles with an inert pharmaceutical diluent and insert inventive composition (see, e.g., U.S. Pat. Nos. 5,916,596 and ing the mixture into a hard gelatin capsule of the appropriate 6,096.331, which are hereby incorporated by reference in size. If soft capsules are desired, a slurry of the nanoparticles their entireties). The following formulations and methods are 15 with an acceptable vegetable oil, light petroleum or other merely exemplary and are in no way limiting. Formulations inert oil can be encapsulated by machine into a gelatin cap Suitable for oral administration can comprise (a) liquid solu Sule. tions, such as an effective amount of the compound dissolved Also provided are unit dosage forms comprising the com in diluents, such as water, saline, or orange juice, (b) capsules, positions and formulations described herein. These unit dos Sachets or tablets, each containing a predetermined amount of age forms can be stored in a Suitable packaging in single or the active ingredient, as solids or granules, (c) Suspensions in multiple unit dosages and may also be further sterilized and an appropriate liquid, (d) Suitable emulsions, and (e) pow sealed. For example, the pharmaceutical composition (e.g., a ders. Tablet forms can include one or more of lactose, man dosage or unit dosage form of a pharmaceutical composition) nitol, corn starch, potato starch, microcrystalline cellulose, may include (i) nanoparticles that comprise rapamycin or a acacia, gelatin, colloidal silicon dioxide, croScarmellose 25 derivative thereof and a carrier protein and (ii) a pharmaceu Sodium, talc, magnesium Stearate, Stearic acid, and other tically acceptable carrier. In other examples, the pharmaceu excipients, colorants, diluents, buffering agents, moistening tical composition (e.g., a dosage or unit dosage form of a agents, preservatives, flavoring agents, and pharmacologi pharmaceutical composition includes a) nanoparticles com cally compatible excipients. Lozenge forms can comprise the prising rapamycin or a derivative thereofanda carrier protein active ingredient in a flavor, usually Sucrose and acacia or 30 (e.g., albumin) and b) at least one other therapeutic agent. In tragacanth, as well as pastilles comprising the active ingredi Some embodiments, the other therapeutic agent comprises a ent in an inert base, such as gelatin and glycerin, or sucrose chemotherapeutic agent (such as any of the chemotherapeutic and acacia, emulsions, gels, and the like containing, in addi agents described herein). In some embodiments, the other tion to the active ingredient, such excipients as are known in therapeutic agent comprises a hormone therapeutic agent. In the art. 35 Some embodiments, the pharmaceutical composition also Formulations suitable for parenteral administration includes one or more other compounds (or pharmaceutically include aqueous and non-aqueous, isotonic sterile injection acceptable salts thereof) that are useful for treating cancer. In Solutions, which can contain anti-oxidants, buffers, bacteri various embodiments, the amount of rapamycin or a deriva ostats, and Solutes that render the formulation compatible tive thereof in the composition is included in any of the with the blood of the intended recipient, and aqueous and 40 following ranges: about 20 to about 50 mg, about 50 to about non-aqueous sterile Suspensions that can include Suspending 100 mg, about 100 to about 125 mg, about 125 to about 150 agents, Solubilizers, thickening agents, stabilizing agents, and mg, about 150 to about 175 mg, about 175 to about 200 mg. preservatives. The formulations can be presented in unit-dose about 200 to about 225 mg, about 225 to about 250 mg, about or multi-dose sealed containers, such as ampules and vials, 250 to about 300 mg. or about 300 to about 350 mg. In some and can be stored in a freeze-dried (lyophilized) condition 45 embodiments, the amount of rapamycin or derivative thereof requiring only the addition of the sterile liquid excipient in the composition (e.g., a dosage or unit dosage form) is in methods of treatment, methods of administration, and dosage the range of about 54 mg to about 540 mg, such as about 180 regimes described herein (i.e., water) for injection, immedi mg to about 270 mg or about 216 mg. of the rapamycin or ately prior to use. Extemporaneous injection Solutions and derivative thereof. In some embodiments, the carrier is suit Suspensions can be prepared from sterile powders, granules, 50 able for parental administration (e.g., intravenous adminis and tablets of the kind previously described. Injectable for tration). In some embodiments, a taxane is not contained in mulations are preferred. the composition. In some embodiments, the rapamycin or Formulations Suitable for aerosol administration comprise derivative thereof is the only pharmaceutically active agent the inventive composition include aqueous and non-aqueous, for the treatment of cancer that is contained in the composi isotonic sterile Solutions, which can contain anti-oxidants, 55 tion. buffers, bacteriostats, and solutes, as well as aqueous and In some embodiments, the invention features a dosage non-aqueous sterile Suspensions that can include Suspending form (e.g., a unit dosage form) for the treatment of cancer agents, Solubilizers, thickening agents, stabilizing agents, and comprising (i) nanoparticles that comprise a carrier protein preservatives, alone or in combination with other suitable and rapamycin or a derivative thereof, wherein the amount of components, which can be made into aerosol formulations to 60 rapamycin or derivative thereof in the unit dosage from is in be administered via inhalation. These aerosol formulations the range of about 180 mg to about 270 mg, and (ii) a phar can be placed into pressurized acceptable propellants, such as maceutically acceptable carrier. In some embodiments, the dichlorodifluoromethane, propane, nitrogen, and the like. amount of the rapamycin or derivative thereof in the unit They also can be formulated as pharmaceuticals for non dosage form includes about 216 mg. pressured preparations, such as in a nebulizer or an atomizer. 65 Also provided are articles of manufacture comprising the In some embodiments, the composition is formulated to compositions, formulations, and unit dosages described have a pH in the range of about 4.5 to about 9.0, including for herein in Suitable packaging for use in the methods of treat US 8,911,786 B2 67 68 ment, methods of administration, and dosage regimes uses further described herein. In some embodiments, the kit described herein. Suitable packaging for compositions of the invention comprises the packaging described above. In described herein are known in the art, and include, for other embodiments, the kit of the invention comprises the example, vials (such as sealed vials), vessels (such as sealed packaging described above and a second packaging compris vessels), ampules, bottles, jars, flexible packaging (e.g., 5 ing a buffer. It may further include other materials desirable sealed Mylar or plastic bags), and the like. These articles of from a commercial and user standpoint, including other buff manufacture may further be sterilized and/or sealed. ers, diluents, filters, needles, Syringes, and package inserts Kits with instructions for performing any methods described The invention also provides kits comprising the composi herein. tions, formulations, unit dosages, and articles of manufacture 10 For combination therapies of the invention, the kit may described hereinforuse in the methods of treatment, methods contain instructions for administering the first and second of administration, and dosage regimes described herein. Kits therapies simultaneously and/or sequentially for the effective of the invention include one or more containers comprising treatment of cancer. The first and second therapies can be rapamycin or a derivative thereof-containing nanoparticle present in separate containers or in a single container. It is compositions (formulations or unit dosage forms and/or 15 understood that the kit may comprise one distinct composi articles of manufacture), and in some embodiments, further tion or two or more compositions wherein one composition comprise instructions for use in accordance with any of the comprises a first therapy and one composition comprises a methods of treatment described herein. In some embodi second therapy. ments, the kit further comprises at least one other therapeutic Kits may also be provided that contain Sufficient dosages of agent. In some embodiments, the other therapeutic agent rapamycin or a derivative thereofas disclosed herein to pro comprises a chemotherapeutic agent (Such as any of the che vide effective treatment for an individual for an extended motherapeutic agents described herein). In some embodi period. Such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 ments, the other therapeutic agent comprises a hormone weeks, 8 weeks, 3 months, 4 months, 5 months, 6 months, 7 therapeutic agent. In some embodiments, the kit comprises i) months, 8 months, 9 months or more. Kits may also include a composition comprising nanoparticles comprising a rapa 25 multiple unit doses of rapamycin or a derivative thereof.com mycin and a carrier protein (such as albumin) and ii) instruc positions, pharmaceutical compositions, and formulations tions for administering the nanoparticles and the chemothera described herein and instructions for use and packaged in peutic agents simultaneously and/or sequentially, for quantities Sufficient for storage and use in pharmacies, for treatment of cancer. In various embodiments, the cancer is example, hospital pharmacies and compounding pharmacies. early stage cancer, non-metastatic cancer, primary cancer, 30 In some embodiments, the kit comprises a dry (e.g., lyo advanced cancer, stage IV cancer, locally advanced cancer, philized) composition that can be reconstituted, resuspended, metastatic cancer, cancer in remission, recurrent cancer, can or rehydrated to form generally a stable aqueous suspension cer in an adjuvant setting, cancer in a neoadjuvant setting, or of nanoparticles comprising rapamycin or a derivative thereof cancer Substantially refractory to hormone treatment. In vari and albumin (e.g., rapamycin or a derivative thereof coated ous embodiments, the amount of rapamycin or a derivative 35 with albumin). thereof in the kit is included in any of the following ranges: The kits of the invention are in Suitable packaging. Suitable about 20 to about 50 mg, about 50 to about 100 mg, about 100 packaging include, but is not limited to, vials, bottles, jars, to about 125 mg, about 125 to about 150 mg, about 150 to flexible packaging (e.g., seled Mylar or plastic bags), and the about 175 mg, about 175 to about 200 mg, about 200 to about like. Kits may optionally provide additional components such 225 mg, about 225 to about 250 mg, about 250 to about 300 40 as buffers and interpretative information. mg, or about 300 to about 350 mg. In some embodiments, the Methods of Making the Compositions amount of rapamycin or a derivative thereof in the kit is in the Methods of making compositions containing carrier pro range of about 54 mg to about 540 mg. Such as about 180 mg teins and poorly water Soluble pharmaceutical agents are to about 270 mg or about 216 mg. In some embodiments, the known in the art. For example, nanoparticles containing kit includes one or more other compounds (i.e., one or more 45 poorly water Soluble pharmaceutical agents and carrier pro compounds other than a taxane) that are useful for treating teins (e.g., albumin) can be prepared under conditions of high cancer. In some embodiments, the other compound is a che shear forces (e.g., Sonication, high pressure homogenization, motherapeutic agent. In some embodiments, the other com or the like). These methods are disclosed in, for example, U.S. pound is a hormone therapeutic. Pat. Nos. 5,916,596; 6,506,405; and 6,537,579 and also in Instructions Supplied in the kits of the invention are typi 50 U.S. Pat. Pub. No. 2005/0004.002A1, which are each hereby cally written instructions on a label or package insert (e.g., a incorporated by reference in their entireties. paper sheet included in the kit), but machine-readable instruc Briefly, the rapamycin or derivative hereof is dissolved in tions (e.g., instructions carried on a magnetic or optical Stor an organic solvent. Suitable organic solvents include, for age disk) are also acceptable. The instructions relating to the example, ketones, esters, ethers, chlorinated solvents, and use of the nanoparticle compositions generally include infor 55 other solvents known in the art. For example, the organic mation as to dosage, dosing schedule, and route of adminis solvent can be methylene chloride, chloroform/ethanol, or tration for the intended treatment. In some embodiments, the chloroform/t-butanol (for example with a ratio of about any of instructions comprise instructions for providing a first and 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, second therapy, wherein either the first or second therapy 7:1.8:1, or 9:1 or with a ratio of about any of 3:7, 5:7, 4:6, 5:5, comprises administering a composition that comprises nano 60 6:5, 8:5, 9:5, 9.5:5, 5:3, 7:3, 6:4, or 9.5:0.5). The solution is particles of rapamycin or derivative thereof and a carrier added to a carrier protein (e.g., human serum albumin). The protein. The kit may further comprise a description of select mixture is Subjected to high pressure homogenization (e.g., ing an individual suitable or treatment. using an Avestin, APV Gaulin, MicrofluidizerTM such as a The present invention also provides kits comprising com MicrofluidizerTM Processor M-1 10EH from Microfluidics, positions (or unit dosages forms and/or articles of manufac 65 Stansted, or Ultra Turrax homogenizer). The emulsion may ture) described herein and may further comprise be cycled through the high pressure homogenizer for between instruction(s) on methods of using the composition, such as about 2 to about 100 cycles, such as about 5 to about 50 cycles US 8,911,786 B2 69 70 or about 8 to about 20 cycles (e.g., about any of 8, 10, 12, 14, second therapy (e.g., one or more other pharmaceutically 16, 18 or 20 cycles). The organic solvent can then be removed active agents for the treatment of cancer). If desired, an anti by evaporation utilizing Suitable equipment known for this microbial agent, Sugar, and/or stabilizing agent can also be purpose, including, but not limited to, rotary evaporators, included in the composition. falling film evaporators, wiped film evaporators, spray driers, Unless defined otherwise, the meanings of all technical and and the like that can be operated in batch mode or in continu Scientific terms used herein are those commonly understood ous operation. The solvent may be removed at reduced pres by one of skill in the art to which this invention belongs. One of skill in the art will also appreciate that any methods and Sure (such as at about any of 25 mm Hg, 30 mm Hg, 40 mm materials similar or equivalent to those described herein can Hg, 50mm Hg, 100 mm Hg, 200 mm Hg, or 300 mmHg). The also be used to practice or test the invention. amount of time used to remove the solvent under reduced 10 The specification is most thoroughly understood in light of pressure may be adjusted based on the volume of the formu the references cited herein. The disclosures of all publica lation. For example, for a formulation produced on a 300 mL tions, patents, patent applications, and published patent appli scale, the solvent can be removed at about 1 to about 300 mm cations referred to herein are each hereby incorporated herein Hg (e.g., about any of 5-100 mm Hg, 10-50 mm Hg, 20-40 by reference in their entireties. mm Hg, or 25 mm Hg) for about 5 to about 60 minutes (e.g., 15 The following Examples are provided to illustrate, but not about any of 7,8,9, 10, 11, 12, 13, 14, 1516, 18, 20, 25, or 30 limit, the invention. minutes). If desired, human albumin solution may be added to the EXAMPLES dispersion to adjust the human serum albumin to rapamycin ratio or to adjust the concentration of rapamycin in the dis The examples, which are intended to be purely exemplary persion. For example, human serum albumin Solution (e.g., of the invention and should therefore not be considered to 25% w/v) can be added to adjust the human serum albuminto limit the invention in any way, also describe and detail aspects rapamycin ratio to about any of 18:1, 15.1 14:1, 13:1, 12:1, and embodiments of the invention discussed above. The 11:1, 10:1, 9:1, 8:1, 7.5:1, 7:1, 6:1, 5:1, 4:1 or 3:1. For examples are not intended to represent that the experiments example, human serum albumin solution (e.g., 25% w/v) or 25 below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used another Solution is added to adjust the concentration of rapa (for example, amounts, temperature, etc.) but some experi mycin in the dispersion to about any of 0.5 mg/ml, 1.3 mg/ml. mental errors and deviations should be accounted for. Unless 1.5 mg/ml, 2 mg/ml, 3 mg/ml. 4 mg/ml, 5 mg/ml, 6 mg/ml, 7 indicated otherwise, parts are parts by weight, molecular mg/ml, 8 mg/ml.9 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 weight is weight average molecular weight, temperature is in mg/ml, 30 mg/ml. 40 mg/ml, or 50 mg/ml. The dispersion 30 degrees Centigrade, and pressure is at or near atmospheric. may be serially filtered through multiple filters, such as a combination of 1.2 Lum and 0.8/0.2um filters; the combination Example 1 of 1.2 m, 0.8 um, 0.45 um, and 0.22 Lum filters; or the combination of any other filters known in the art. The disper Exemplary Methods for the Formation of sion obtained can be further lyophilized. The nanoparticle 35 Nanoparticle Compositions with Rapamycin and compositions may be made using a batch process or a con Albumin tinuous process (e.g., the production of a composition on a large Scale). Example 1-A If desired, a second therapy (e.g., one or more compounds useful for treating breast cancer), an antimicrobial agent, 40 This example demonstrates the preparation of a pharma Sugar, and/or stabilizing agent can also be included in the ceutical composition comprising rapamycin and albumin in composition. This additional agent can either be admixed which the rapamycin concentration was 8 mg/mL in the emul with the rapamycin and/or the carrier protein during prepa sion and the formulation was made on a 300 mL scale. Rapa mycin (2400 mg) was dissolved in 12 mL of chloroform/t- ration of the rapamycin/carrier protein composition, or added butanol. The solution was then added into 288 mL of a human after the rapamycin/carrier protein composition is prepared. 45 serum albumin solution (3% w/v). The mixture was homog For example, the agent can be added along with an aqueous enized for 5 minutes at 10,000 rpm (Vitris homogenizer medium used to reconstitute? suspend the rapamycin/carrier model Tempest I.Q.) in order to form a crude emulsion, and protein composition or added to an aqueous Suspension of the then transferred into a high pressure homogenizer. The emul carrier protein-associated rapamycin. In some embodiments, sification was performed at 20,000 psi while recycling the the agent is admixed with the rapamycin/carrier protein com 50 emulsion. The resulting system was transferred into a position prior to lyophilization. In some embodiments, the Rotavap, and the solvent was rapidly removed at 40° C. at agent is added to the lyophilized pharmaceutical agent/carrier reduced pressure (25 mm of Hg). The resulting dispersion protein composition. In some embodiments when the addi was translucent. At this stage, human serum albumin Solution tion of the agent changes the pH of the composition, the pH in was added to the dispersion to adjust the human serum albu the composition are generally (but not necessarily) adjusted 55 minto rapamycin ratio. The dispersion was serially filtered to a desired pH. Exemplary pH values of the compositions through multiple filters. The size of the filtered formulation include, for example, in the range of about 5 to about 8.5. In was 85-100 nm (Z. Malvern Zetasizer). The dispersion was Some embodiments, the pH of the composition is adjusted to further lyophilized (FTS Systems. Dura-Dry LP, Stone Ridge, no less than about 6, including for example no less than any of N.Y.) for 60 hours. The resulting cake was easily reconstitut about 6.5, 7, or 8 (e.g., about 8). 60 able to the original dispersion by the addition of sterile water The invention also provides methods of making the com or 0.9% (w/v) sterile saline. The particle size after reconsti bination therapies described herein for use in the treatment of tution was the same as before lyophilization. cancer. For example, there is provided a method of preparing a composition comprising rapamycin or a derivative thereof, Example 1-B a carrier protein (e.g., albumin), and a second therapy by 65 combining (e.g., admixing) a composition containing rapa This example demonstrates the preparation of a pharma mycin (or a derivative thereof) and a carrier protein with a ceutical composition comprising rapamycin and albumin in US 8,911,786 B2 71 72 which the rapamycin concentration was 8.3 mg/mL in the nm (Z. Malvern Zetasizer). The liquid Suspension was emulsion and the formulation was made on a 200 mL scale. found to be stable at 4° C. and 25° C. at least for 48 hours. Rapamycin (1660 mg) was dissolved in 8.5 mL of chloro form/ethanol. The solution was then added into 191.5 mL of Example 1-E a human serum albumin solution (6% w/v). The mixture was 5 homogenized for 5 minutes at 10,000 rpm (Vitris homog This example demonstrates the preparation of a pharma enizer model Tempest I.Q.) in order to form a crude emulsion, ceutical composition comprising rapamycin and albumin in and then transferred into a high pressure homogenizer. The which the rapamycin concentration was 8.5 mg/mL in the emulsification was performed at 20,000 psi while recycling emulsion and the formulation was made on a 30 mL scale. the emulsion. The resulting system was transferred into a 10 Rapamycin (255 mg) was dissolved in 1.35 mL of chloro Rotavap, and the solvent was rapidly removed at 40° C. at form/ethanol. The solution was then added into 28.7 mL of a reduced pressure (25 mm of Hg). The dispersion was serially human serum albumin solution (6% w/v). The mixture was filtered. The size of the 0.22 um filtered formulation was 85 homogenized for 5 minutes at 10,000 rpm (Vitris homog nm (Z. Malvern Zetasizer). The dispersion was further lyo enizer model Tempest I.Q.) in order to form a crude emulsion, philized (FTS Systems. Dura-Dry LP, Stone Ridge, N.Y.) for 15 and then transferred into a high pressure homogenizer. The emulsification was performed at 20,000 psi while recycling 60 hours. The resulting cake was easily reconstitutable to the the emulsion. The resulting system was transferred into a original dispersion by addition of 0.9% (w/v) sterile saline. Rotavap, and the solvent was rapidly removed at 40° C. at The particle size after reconstitution was the same as before reduced pressure (40 mm of Hg). The dispersion was serially lyophilization. filtered. The size of the 0.22 Lum filtered formulation was 136 nm (Z. Malvern Zetasizer). The liquid Suspension was Example 1-C found to be stable at 4° C. and 25° C. at least for 24 hours. This example demonstrates the preparation of a pharma Example 1-F ceutical composition comprising rapamycin and albumin in 25 which the rapamycin concentration was 16.2 mg/mL in the This example demonstrates the preparation of a pharma emulsion and the formulation was made on a 200 mL scale. ceutical composition comprising rapamycin and albumin in Rapamycin (3240 mg) was dissolved in 16 mL of chloroform/ which the rapamycin concentration was 9.2 mg/mL in the ethanol. The solution was then added into 184 mL of a human emulsion and the formulation was made on a 20 mL scale. serum albumin solution (6% w/v). The mixture was homog 30 Rapamycin (184 mg) was dissolved in 1.0 mL of chloroform/ enized for 5 minutes at 10,000 rpm (Vitris homogenizer ethanol. The solution was then added into 19.0 mL of a human model Tempest I.Q.) in order to form a crude emulsion, and serum albumin solution (7% w/v). The mixture was homog then transferred into a high pressure homogenizer. The emul enized for 5 minutes at 10,000 rpm (Vitris homogenizer sification was performed at 20,000 psi while recycling the model Tempest I.Q.) in order to form a crude emulsion, and emulsion. The resulting system was transferred into a 35 then transferred into a high pressure homogenizer. The emul Rotavap, and the solvent was rapidly removed at 40° C. at sification was performed at 20,000 psi while recycling the emulsion. The resulting system was transferred into a reduced pressure (25 mm of Hg). At this stage, human serum Rotavap, and the solvent was rapidly removed at 40° C. at albumin solution was added to the dispersion and the volume reduced pressure (40 mm of Hg). The dispersion was serially of the dispersion was made to 400 mL to adjust the human 40 filtered. The size of the 0.22 Lum filtered formulation was 124 serum albuminto rapamycin ratio and to adjust the rapamycin nm (Z. Malvern Zetasizer). The liquid Suspension was concentration. The dispersion was serially filtered. The size found to be stable at 4° C. and 25° C. at least for 24 hours. of the 0.22 um filtered formulation was 99 nm (Z. Malvern Zetasizer). The dispersion was further lyophilized (FTS Sys Example 1-G tems. Dura-Dry LP, Stone Ridge, N.Y.) for 60 hours. The 45 resulting cake was easily reconstitutable to the original dis This example demonstrates the preparation of a pharma persion by addition of 0.9% (w/v) sterile saline. The particle ceutical composition comprising rapamycin and albumin in size after reconstitution was the same as before lyophiliza which the rapamycin concentration was 8.4 mg/mL in the tion. emulsion and the formulation was made on a 20 mL scale. 50 Rapamycin (168 mg) was dissolved in 1.2 mL of chloroform/ Example 1-D ethanol. The solution was then added into 18.8 mL of a human serum albumin solution (6% w/v). The mixture was homog This example demonstrates the preparation of a pharma enized for 5 minutes at 10,000 rpm (Vitris homogenizer ceutical composition comprising rapamycin and albumin in model Tempest I.Q.) in order to form a crude emulsion, and which the rapamycin concentration was 8.2 mg/mL in the 55 then transferred into a high pressure homogenizer. The emul emulsion and the formulation was made on a 40 mL scale. sification was performed at 20,000 psi while recycling the Rapamycin (328 mg) was dissolved in 1.8 mL of chloroform/ emulsion. The resulting system was transferred into a ethanol. The solution was then added into 38.2 mL of a human Rotavap, and the solvent was rapidly removed at 40° C. at serum albumin solution (6% w/v). The mixture was homog reduced pressure (40 mm of Hg). The dispersion was serially enized for 5 minutes at 10,000 rpm (Vitris homogenizer 60 filtered. The size of the 0.22 um filtered formulation was 95 model Tempest I.Q.) in order to form a crude emulsion, and nm (Z.e Malvern Zetasizer). then transferred into a high pressure homogenizer. The emul sification was performed at 20,000 psi while recycling the Example 1-H emulsion. The resulting system was transferred into a Rotavap, and the solvent was rapidly removed at 40° C. at 65 This example demonstrates the preparation of a pharma reduced pressure (40 mm of Hg). The dispersion was serially ceutical composition comprising rapamycin and albumin in filtered. The size of the 0.22 Lum filtered formulation was 108 which the rapamycin concentration was 8.2 mg/mL in the US 8,911,786 B2 73 74 emulsion and the formulation was made on a 20 mL scale. reduced pressure (40 mm of Hg). The dispersion was serially Rapamycin (164 mg) was dissolved in 0.9 mL of chloroform/ filtered. The size of the 0.22 um filtered formulation was 99 ethanol. The solution was then added into 19.1 mL of a human nm (Z.: Malvern Zetasizer). serum albumin solution (8% w/v). The mixture was homog enized for 5 minutes at 10,000 rpm (Vitris homogenizer 5 Example 1-L model Tempest I.Q.) in order to form a crude emulsion, and then transferred into a high pressure homogenizer. The emul This example demonstrates the preparation of a pharma sification was performed at 20,000 psi while recycling the ceutical composition comprising rapamycin and albumin in emulsion. The resulting system was transferred into a which the rapamycin concentration was 5.0 mg/mL in the Rotavap, and the solvent was rapidly removed at 40° C. at 10 emulsion and the formulation was made on a 20 mL scale. reduced pressure (40 mm of Hg). The dispersion was serially Rapamycin (100 mg) was dissolved in 0.8 mL of chloroform/ filtered. The size of the 0.22 Lum filtered formulation was 149 ethanol. The solution was then added into 19.2 mL of a human nm (Z.: Malvern Zetasizer). serum albumin solution (3% w/v). The mixture was homog enized for 5 minutes at 10,000 rpm (Vitris homogenizer Example 1-I 15 model Tempest I.Q.) in order to form a crude emulsion, and then transferred into a high pressure homogenizer. The emul This example demonstrates the preparation of a pharma sification was performed at 20,000 psi while recycling the ceutical composition comprising rapamycin and albumin in emulsion. The resulting system was transferred into a which the rapamycin concentration was 6.6 mg/mL in the Rotavap, and the solvent was rapidly removed at 40° C. at emulsion and the formulation was made on a 20 mL scale. reduced pressure (40 mm of Hg). The dispersion was serially Rapamycin (132 mg) was dissolved in 0.8 mL of chloroform/ filtered. The size of the 0.22 Lum filtered formulation was 146 ethanol. The solution was then added into 19.2 mL of a human nm (Z.e Malvern Zetasizer). serum albumin solution (5% w/v). The mixture was homog enized for 5 minutes at 10,000 rpm (Vitris homogenizer Example 1-M model Tempest I.Q.) in order to form a crude emulsion, and 25 then transferred into a high pressure homogenizer. The emul This example demonstrates the preparation of a pharma sification was performed at 20,000 psi while recycling the ceutical composition comprising rapamycin and albumin in emulsion. The resulting system was transferred into a which the rapamycin concentration was 4.0 mg/mL in the Rotavap, and the solvent was rapidly removed at 40° C. at emulsion and the formulation was made on a 20 mL scale. reduced pressure (40 mm of Hg). The dispersion was serially 30 Rapamycin (80 mg) was dissolved in 0.8 mL of chloroform/ filtered. The size of the 0.22 Lum filtered formulation was 129 ethanol. The solution was then added into 19.2 mL of a human nm (Z.: Malvern Zetasizer). serum albumin solution (3% w/v). The mixture was homog enized for 5 minutes at 10,000 rpm (Vitris homogenizer Example 1-J model Tempest I.Q.) in order to form a crude emulsion, and 35 then transferred into a high pressure homogenizer. The emul This example demonstrates the preparation of a pharma sification was performed at 20,000 psi while recycling the ceutical composition comprising rapamycin and albumin in emulsion. The resulting system was transferred into a which the rapamycin concentration was 4.0 mg/mL in the Rotavap, and the solvent was rapidly removed at 40° C. at emulsion and the formulation was made on a 20 mL scale. reduced pressure (40 mm of Hg). The resulting dispersion Rapamycin (80 mg) was dissolved in 0.8 mL of chloroform/ 40 was a white milky Suspension. The dispersion was serially ethanol. The solution was then added into 19.2 mL of a human filtered. The size of the 0.22 Lum filtered formulation was 129 serum albumin solution (3% w/v). The mixture was homog nm (Z.: Malvern Zetasizer). enized for 5 minutes at 10,000 rpm (Vitris homogenizer model Tempest I.Q.) in order to form a crude emulsion, and Example 1-N then transferred into a high pressure homogenizer. The emul 45 sification was performed at 20,000 psi while recycling the This example demonstrates the preparation of a pharma emulsion. The resulting system was transferred into a ceutical composition comprising rapamycin and albumin in Rotavap, and the solvent was rapidly removed at 40° C. at which the rapamycin concentration was 4.0 mg/mL in the reduced pressure (40 mm of Hg). The dispersion was serially emulsion and the formulation was made on a 20 mL scale. filtered. The size of the 0.22 Lum filtered formulation was 108 50 Rapamycin (80 mg) was dissolved in 0.8 mL of chloroform/ nm (Z.: Malvern Zetasizer). ethanol. The solution was then added into 19.2 mL of a human serum albumin solution (3% w/v). The mixture was homog Example 1-K enized for 5 minutes at 10,000 rpm (Vitris homogenizer model Tempest I.Q.) in order to form a crude emulsion, and This example demonstrates the preparation of a pharma 55 then transferred into a high pressure homogenizer. The emul ceutical composition comprising rapamycin and albumin in sification was performed at 20,000 psi while recycling the which the rapamycin concentration was 4.0 mg/mL in the emulsion. The resulting system was transferred into a emulsion and the formulation was made on a 20 mL scale. Rotavap, and the solvent was rapidly removed at 40° C. at Rapamycin (80 mg) was dissolved in 0.8 mL of chloroform/ reduced pressure (40 mm of Hg). The dispersion was serially ethanol. The solution was then added into 192 mL of a human 60 filtered. The size of the 0.22 Lum filtered formulation was 166 serum albumin solution (1% w/v). The mixture was homog nm (Z.e Malvern Zetasizer). enized for 5 minutes at 10,000 rpm (Vitris homogenizer model Tempest I.Q.) in order to form a crude emulsion, and Example 1-O then transferred into a high pressure homogenizer. The emul sification was performed at 20,000 psi while recycling the 65 This example demonstrates the preparation of a pharma emulsion. The resulting system was transferred into a ceutical composition comprising rapamycin and albumin in Rotavap, and the solvent was rapidly removed at 40° C. at which the rapamycin concentration was 4.0 mg/mL in the US 8,911,786 B2 75 76 emulsion and the formulation was made on a 20 mL scale. Nab-rapamycin used were 0, 15, 30, 45, 90 and 180 mg/kg Rapamycin (80 mg) was dissolved in 0.8 mL of chloroform/ with a q4dx3 schedule. The pharmacokinetics of Nab-rapa ethanol. The solution was then added into 19.2 mL of a human mycin was also investigated in Sprague Dawley rats at dose serum albumin solution (3% w/v). The mixture was homog levels of 1 (N=3), 15 (N=4), 30 (N=3), and 45 mg/kg (N=4). enized for 5 minutes at 10,000 rpm (Vitris homogenizer 5 model Tempest I.Q.) in order to form a crude emulsion, and Blood samples were collected prior to dosing (baseline) and then transferred into a high pressure homogenizer. The emul post-dosing at the following time points: 1, 5, 10, 15, 30 and sification was performed at 20,000 psi while recycling the 45 minutes, and 1, 4, 8, 24, 36 and 48 hours. Plasma samples emulsion. The resulting system was transferred into a were analyzed for rapamycin using LC/MS. Rotavap, and the solvent was rapidly removed at 40° C. at Nab-rapamycin was nontoxic at the highest dose of 180 reduced pressure (40 mm of Hg). The dispersion was serially 10 mg/kg on a q4dX3 schedule. No changes in blood chemistry filtered. The size of the 0.22 um filtered formulation was 90 or CBC were observed. No hypercholesterolemia and hyper nm (Z.: Malvern Zetasizer). triglyceridemia were observed. As illustrated in FIGS. 1 and 2C, Nab-rapamycin exhibited linear pharmacokinetics with Example 1-P respect to dose and rapid extravascular distribution as dem 15 onstrated by large Vss and VZ. The Cmax and AUCinf of This example demonstrates the preparation of a pharma Nab-rapamycin were dose proportional (FIGS. 2A and 2B, ceutical composition comprising rapamycin and albumin in respectively). which the rapamycin concentration was 4.0 mg/mL in the If desired, other compositions of the invention (e.g., com emulsion and the formulation was made on a 20 mL scale. positions that contain rapamycin derivatives or carrier pro Rapamycin (80 mg) was dissolved in 0.8 mL of chloroform/ teins other than human serum albumin) can be tested in these ethanol. The solution was then added into 19.2 mL of a human assays for toxicity and pharmacokinetics. serum albumin solution (3% w/v). The mixture was homog enized for 5 minutes at 10,000 rpm (Vitris homogenizer Example 2B model Tempest I.Q.) in order to form a crude emulsion, and then transferred into a high pressure homogenizer. The emul 25 Toxicology and Pharmacokinetic Studies of sification was performed at 20,000 psi while recycling the Nab-rapamycin emulsion. The resulting system was transferred into a Rotavap, and the solvent was rapidly removed at 40° C. at reduced pressure (40 mm of Hg). The dispersion was serially The overall toxicity of Nab-rapamycin was determined in a filtered. The size of the 0.22 um filtered formulation was 81 dose ranging study in Sprague Dawley rats. Nab-rapamycin 30 was intravenously administered at 0, 20, 40,90, 120, and 180 nm (Z.: Malvern Zetasizer). mg/kg on a q4dX3 schedule on days 1, 5, and 9 (n=20). Example 1-Q Nab-rapamycin was well tolerated at dose levels up to 90 mg/kg (540 mg/m) on a q4.dx3 schedule. There was 20% and This example demonstrates the preparation of a pharma 100% mortality among the highest doses of 120 mg/kg and ceutical composition comprising rapamycin and albumin. 35 180 mg/kg. No hypercholesterolemia and hypertriglyceri Rapamycin (30 mg) was dissolved in 2 ml chloroform/etha demia were observed. nol. The solution was then added into 27.0 ml of a human The pharmacokinetics of Nab-rapamycin was also investi serum albumin solution (3% w/v). The mixture was homog gated in Sprague Dawley rats at dose levels of 1 (N=5), 15 enized for 5 minutes at low RPM (Vitris homogenizer model (N=4), 30 (N=3), and 45 mg/kg (N=4). Blood samples wec Tempest I.Q.) in order to form a crude emulsion, and then 40 ollected prior to dosing (baseline) and post-dosing at the transferred into a high pressure homogenizer. The emulsifi following time points: 1, 5, 10, 15, 30 and 45 minutes, and 1. cation was performed at 9000-40,000 psi while recycling the 4, 8, 24, 36 and 48 hours. Plasma samples were analyzed for emulsion for at least 5 cycles. The resulting system was rapamycin using LC/MS. transferred into a Rotavap, and the solvent was rapidly Nab-rapamycin exhibited a very rapid distribution phase removed at 40°C. at reduced pressure (30 mm Hg) for 20-30 45 and large V-andVss. The C, and AUC of Nab-rapamycin minutes. The resulting dispersion was translucent, and the were dose proportional. See FIG.1. The PK of Nab-rapamyin typical average diameter of the resulting particles was in the is similar to Nab-paclitaxel and Nab-docetaxel. FIG. 2D range 50-220 nm (Z-average, Malvern Zetasizer). The disper shows the log-linerar plot Nab-rapamycin blood concentra sion was further lyophilized for 48 hours. The resulting cake tion vs. time following IV administration to rats at dose levels was easily reconstituted to the original dispersion by addition 50 of 15 mg/kg, 30 mg/kg, and 45 mg/kg. ofsterile water or saline. The particle size after reconstitution was the same as before lyophilization. Example 3 If desired, other compositions of the invention (e.g., com positions that contain rapamycin derivatives or carrier pro Inhibition of Breast Cancer Cells Using teins other than human serum albumin) can be made using 55 Nab-rapamycin these methods or a variation of these methods. It should be recognized that the amounts, types, and proportions of drug, The antitumor activity of Nab-rapamycin was examined Solvents, and proteins used in these examples are not limiting using a human mammary carcinoma xenograft in mice. MX-1 in any way. tumors were implanted Subcutaneously into both the right and 60 left flanks of female athymic mice (4-5 per group) and Example 2A allowed to grow to 100 mm. The mice were then intrave nously administered either saline or Nab-rapamycin at a dose Toxicology and Pharmacokinetic Studies of level of 40 mg/kg with a three times weekly schedule for 4 Nab-rapamycin weeks. The dosing Volume was 2 ml/kg. Tumor growth data 65 were analyzed by ANOVA. The overall toxicity of Nab-rapamycin was determined in a Nab-rapamycin was highly effective against breast cancer, dose ranging study in Sprague Dawley rats. The dose levels of achieving a tumor growth inhibition of 88% against the MX-1 US 8,911,786 B2 77 78 xenograft (p<0.0001 versus control, ANOVA: FIG. 3A). No For these cell-based assays, RPMI 8226 and U266 human significant weight loss was observed in the mice from Nab MM cell lines are obtained from the American Type Culture rapamycin at 40 mg/kg (FIG.3B). Thus, Nab-rapamycin was Collection (ATCC) of Rockville, Md. Patient derived MM well tolerated even at the highest dose of 180 mg/kg with a cells are purified from patient BMsamples, as described by Y. T. Tai, G. Teoh, Y. Shima, et al., J. Immunol. Methods 235:11, q4dX3 schedule, showed linear pharmacokinetics, and was 2000. Human MM cell lines are cultured in RPMI-1640 highly effective against a breast cancer model in vivo. media (Sigma Chemical, St. Louis, Mo.), containing 10% If desired, other compositions of the invention (e.g., com fetal bovine serum (FBS), 2 mmol/L L-glutamine (L-glut, positions that contain rapamycin derivatives or carrier pro GIBCO, Grand Island, N.Y.), 100 U/mL penicillin and 100 teins other than human serum albumin) can be tested in this mg/mL streptomycin (P/S, GIBCO). MM patient cells are animal model to determine their ability to treat breast cancer 10 95% CD38+, CD45RA-. Bone marrow stromal cells (BM in vivo. SCs) are prepared from aspirates of MM patients as well as healthy donors as described by D. Gupta, S. Treon, Y. Shima, Example 4 et al. in Leukemia, 2001 and S. Gartner and H. S. Kaplan in Proc. Nag. Acad. Sci. USA 77:4756, 1980. Cells are cultured Use of Human Clinical Trials to Determine the 15 in ISCOVE's modified Dulbecco media containing 20% FBS, 2 mmol/L L-glut, and 100 ug/mLP/S. Human umbilical Ability of Compositions of the Invention to Treat, vein endothelial cells (HUVEC P168) are purchased from Stabilize, Prevent, and/or Delay Cancer Clonetics, Biowhittaker, and maintained in EGM-2MV media (Clonetics, Biowhittaker). The nanoparticles compris If desired, any of the compositions described herein can ing rapamycin and a carrier protein (such as albumin) are also be tested in humans to determine the ability of the com diluted in culture medium to concentrations ranging, e.g., positions to treat, stabilize, prevent and/or delay cancer (e.g., from 0.01 to 100 uM. breast cancer). Standard methods can be used for these clini cal trials. Example 6 In one exemplary method, Subjects (e.g., healthy Subjects, 25 Subjects with cancer Such as breast cancer, or Subjects at Panel of Drug-Resistant MMCell Lines and Primary increased risk for cancer Such as breast cancer) are enrolled in MMTumor Cells for Use in Determination of a tolerability, pharmacokinetics, and pharmacodynamics Nab-rapamycin Activity phase I study of Nab-rapamycin or a derivative thereofusing Effectiveness of the nanoparticle compositions of the standard protocols. For example, escalating doses of rapamy 30 invention may further be evaluated in drug resistant cell lines. cin or a derivative thereof up to about 250 mg/m as part of a The use of drug resistant cells facilitates the determination of composition of the invention can be tested. Then a phase II, potential cancer patient Subpopulations that may be effec double-blind randomized controlled trial is performed to tively treated by the use of the nanoparticle compositions of determine the efficacy of the Nab-rapamycin or a derivative the invention. The activity of any of the nanoparticle compo thereof If desired, the activity of Nab-rapamycin or a deriva 35 sitions of the invention (e.g., nanoparticles comprising rapa tive thereof can be compared to that of another treatment for mycin and a carrier protein Such as albumin) can be evaluated cancer (e.g., breast cancer). Alternatively or additionally, the in a panel of drug-sensitive and drug-resistant human MM efficacy of a combination of Nab-rapamycin or a derivative cell lines using standard methods. Exemplary cell lines thereof and another treatment for cancer (e.g., breast cancer) include a dexamethasone (Dex)-sensitive MM-1S cell line, a can be compared to that of either treatment alone. 40 Dex-resistant MM-1R cell line; the chemo-sensitive parental MM cell line RPMI-8226/S, and its chemo-resistant Sublines Example 5 RPMI-8226/Dox40 (doxorubicin-resistant), RPMI-8226/ MR20 (mitoxantrone-resistant), and RPMI-8226/LR5 (mel Multiple Myeloma (MM) Cell Lines for Use in phalan-resistant) cells; MM-1S-TR15 is a TRAIL/Apo2L Determination of Nab-rapamycin Activity 45 resistant subline: MM-SAR-1 (also referred to as MM-SA-1) cells that are primary MM tumor cells from a patient resistant to the proteasome inhibitor bortezomib (PS-341) (cells main Interleukin-6 (IL-6) and insulin like growth factor-1 (IGF tained in vitro resistance to PS-341):OCI-My-5 cells; S6B45 1) play a key role in the growth, Survival, and drug resistance cells; ARD; ARK; ARP-1 OPM-1: OPM-6: K620; LP-1: in multiple myeloma (MM) cells. Furthermore, their secre U266; and NCI-H929 cells. All cells are cultured in RPMI tion in bone marrow stromal cells (BMSCs) is up-regulated 50 1640 medium (Life Technologies, Grand Island, N.Y.) by adherence of MM cells. IL-6 and IGF-1 mediate growth of supplemented with 10% fetal bovine serum, L-glutamine, MM cells via activation of the mitogen-activated protein penicillin, and streptomycin (Life Technologies). kinase (MAPK) and phosphatidylinositol 3'-kinase/Akt Primary MMtumor cells additionally may be isolated from kinase (PI3-K/Akt) signaling cascades. Several Studies show bone marrow (BM) aspirates of patients, who are resistant to that PI3-K/Akt signaling mediates growth, Survival, migra 55 conventional (steroid- and cytotoxic chemotherapy-based) tion and cell cycle regulation in MM. Activated Akt in turn and more recently developed anti-MM agents (e.g. thalido phosphorylates downstream target molecules, including mide or proteasome inhibitors). The resistant primary MM forkhead transcription factor (FKHR), glycogen synthase tumor cells are collected from patients as described above in kinase (GSK)-3B, and mammalian target of rapamycin Example 4. (mTOR). 60 MM cell lines can be used in standard cell-based assays to Example 7 test the ability of any of the nanoparticle compositions of the invention (e.g., nanoparticles comprising rapamycin and a Co-Culture Assays of MMCells with Bone Marrow carrier protein such as albumin) to treat MM. The nanopar Stromal Cells (BMSCs) Treated with Nab-rapamycin ticle compositions of the invention are desirable because they 65 may allow rapamycin to be delivered at higher doses with When adhering to BMSCs, MM cells have reduced sensi improved efficacy. tivity to conventional anti-MM therapies, such as dexametha US 8,911,786 B2 79 80 Sone or cytotoxic chemotherapeutics (Chauhan D. et al., media containing 10% fetal bovine serum, and then plated Blood. 1996, 87, 1104-1112). This form of drug resistance is into 96-well microtiter plates (Costar, Cambridge, Mass.), in considered a key reason why MM patients eventually relapse the presence of a nanoparticle composition of the invention when they receive treatment based on administration of glu (e.g., nanoparticles comprising rapamycin and a carrier pro cocorticoids and/or cytotoxic chemotherapy. Therefore, any tein such as albumin) or DMSO control. Proliferation is mea of the nanoparticle compositions of the invention (e.g., nano sured by the incorporation of H-thymidine (NEN Products, particles comprising rapamycin and a carrier protein Such as Boston, Mass.). Specifically, cells are pulsed with H-thymi albumin) can be tested to determine whether they overcome dine (0.5 muck?well) for the last 6 hours of 48 hour cultures, the molecular sequelae of the interaction of BMSCs with MM harvested onto glass filters with an automatic cell harvester cells and achieve anti-MM activity in this context. In particu 10 (Cambridge Technology, Cambridge, Mass.), and counted lar, an in vitro co-culture assay is performed using MM cells using a LKB Betaplate scintillation counter (Wallac, Gaith with BMSCs as previously described. BMSCs are grown on ersburg, Md.). Measurement of cell viability is performed 24-well plates to confluency. Following washings with colorimetrically using a MTS assay, utilizing the CelTiter96 serum-free medium, primary tumor cells (greater than about One Solution Reagent (Promega, Madison, Wis.). Cells are 95% purity in CD138+ cells) isolated from MM patients are 15 exposed to the MTS for the last 2 hours of 48 hour cultures, added to BMSC-coated or control wells as described previ and absorbance is measured using an ELISA plate reader ously (Uchiyama H. et al., Blood 1993, 82,3712-3720; Mit (Molecular Devices Corp., Sunnyvale, Calif.) at OD of 570 siades N. et al., Blood 2003, 101, 4055-4062) and incubated . for 48 hours in the presence or absence of a nanoparticle composition of the invention, such as nab-rapamycin. Flow Example 10 cytometric analysis is performed to detect the CD138+ popu lation of viable MM cells and the effect of the nanoparticle Cell Cycle Analysis of MM Tissue Culture Cells composition on MM cell viability is expressed as a percent of Treated with Nab-rapamycin viable cells in comparison to the respective vehicle-treated cultures. 25 In this example, the effect of nanoparticle composition of the invention (e.g., nanoparticles comprising rapamycin and a Example 8 carrier protein such as albumin) on cell cycle is assessed. MM cells (1x106 cells) are cultured in the presence of a nanopar MTT Calorimetric Survival Assay of MM Tissue ticle composition of the invention (e.g., nanoparticles com Culture Cells Treated with Nab-rapamycin 30 prising rapamycin and a carrier protein Such as albumin) or DMSO control for 24, 48 and 72 hours. Cells are thenwashed In this example, the effect of nanoparticle composition of with phosphate buffered saline (PBS), fixed with 70% etha the invention (e.g., nanoparticles comprising rapamycin and a nol, and treated with RNAse (Sigma). Cells are next stained carrier protein such as albumin) on cell viability and survival with propidium iodide (PI, 5ug/mL), and the cell cycle profile is assessed. Cell Survival is examined using a 3-(4,5-dimeth 35 is determined using the M software on an Epics flow cytom ylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT; eter (Coulter Immunology, Hialeah, Fla.). Sigma Chemical, St Louis, Mo.) colorimetric assay, as pre viously described (Mitsiades C. S. et al., Blood 2001, 98, Example 11 795-804; Mitsiades N. et al., PNAS 2002, 99, 14374-14379; Mitsiades N. et al., Blood 2003, 101, 2377-2380). Briefly, 40 Other MM Cell Activity Assays for Cells Treated cells are plated in 48-well plates at 70% to 80% confluence in with Nab-rapamycin the presence of 2.5% fetal bovine serum (FBS) and in the presence of a nanoparticle composition of the invention (e.g., Nanoparticle composition of the invention (e.g., nanopar nanoparticles comprising rapamycin and a carrier protein ticles comprising rapamycin and a carrier protein Such as such as albumin) at final concentration of 0-100 nM rapamy 45 albumin) can be further assessed by other activity assays cin or DMSO vehicle control. At the end of each treatment, known in the art. For example, the molecular mechanisms of cells are incubated with 1 mg/mL MTT for 4 hours at 37°C. anti-MM activities of nanoparticle composition of the inven A mixture ofisopropanol and 1 NHCl (23:2, Vol/vol) is then tion may be assessed using, but not limited to, cell cycle added under vigorous pipetting to dissolve the formazan crys profiling by caspases/PARP cleavage and quantification of tals. Dye absorbance (A) in viable cells is measured at 570 50 anti-apoptotic proteins by Western blotting. nm, with 630 nm as a reference wavelength. Cell viability is estimated as a percentage of the value of untreated controls. Example 12A Experiments are typically repeated at least 3 times, and each experimental condition is typically repeated at least in tripli Effect of Nab-rapamycin on Human MMCells in cate wells in each experiment. Data is reported are average 55 vivo values+/-SD of representative experiments. In this example, the effect of nanoparticle composition of Example 9 the invention (e.g., nanoparticles comprising rapamycin and a carrier protein such as albumin) on MM cell growth in vivo is Proliferation of MMCells Treated with 60 assessed. Mice are inoculated Subcutaneously into the right Nab-rapamycin flank with 3x107 MM cells in 100 mL of RPMI 1640, together with 100 uL matrigel basement membrane matrix In this example, the effect of nanoparticle composition of (Becton Dickinson, Bedford, Mass.). On day 6 post injection, the invention (e.g., nanoparticles comprising rapamycin and a mice areassigned into two treatment groups receiving a nano carrier protein Such as albumin) on cell proliferation and 65 particle composition of the invention (e.g., nanoparticles viability is assessed. For proliferation and cell viability comprising rapamycin and a carrier protein such as albumin) assays, MM cells are first starved for 12 hours in RPMI-1640 or into a control group. Treatment with a nanoparticle com US 8,911,786 B2 81 82 position of the invention is then intravenously administered DMSO at 2 mL/kg or Nab-rapamycin at a dose level of 40 either saline or nanoparticles comprising rapamycin and a mg/kg with a three times weekly schedule for 4 weeks at a carrier protein such as albumin at a dose level of 40 mg/kg dosing Volume of 5 mL/kg. Tumor growth data were analyzed with a three times weekly schedule for 4 weeks. The dosing by ANOVA. Volume is 2 ml/kg. Caliper measurements of the longest per Nab-rapamycin significantly inhibited in vivo tumor pendicular tumor diameters are performed twice per week to growth for HT29 tumors, achieving a tumor growth inhibition estimate the tumor volume. Animals are sacrificed when their of 78.9% against the HT29 tumor xenograft (p=0.005 versus tumor reached 2 cm or when the mice become moribund. control, ANOVA: FIG. 5A). A -9.2% weight loss was Survival is evaluated from the first day of tumor injection observed in the mice from Nab-rapamycin at 40 mg/kg (FIG. until death. 10 5B). Example 12B Example 15 Effect of Nab-rapamycin on Human MM1S Cells in Cytotoxic Activity of Nab-rapamycin Against 15 HCT-116 (Human Colon Carcinoma) Tumor vivo Xenograft In this example, the effect of Nab-rapamycin on MM cell growth in vivo was assessed. Mice were inoculated Subcuta The antitumor activity of Nab-rapamycin was examined neously into the right flank with 3x107 MM1S cells in 100 mL using HCT-116 human colon carcinoma xenograft in mice. of RPMI 1640, together with 100LL matrigelbasement mem HCT-116 tumors were implanted subcutaneously into the brane matrix (Becton Dickinson, Bedford, Mass.). On day 6 right flanks of male athymic nude mice (10 per group) and post injection, mice were assigned into three treatment groups allowed to grow to 100-221 mm. The mice were then intra receiving Nab-rapamycin or into a control group. Animals in venously administered with either saline or Nab-rapamycinat the control group were administered with 0.9% NaCl solution a dose level of 40 mg/kg with a three times weekly schedule (i.v.). Animals in the three treatment groups were adminis 25 for 4 weeks at a dosing Volume of 10 mL/kg. Tumor growth tered with Nab-rapamycin at a dose schedule of 20 or 40 data were analyzed by ANOVA. mg/kg three times weekly or at a dose schedule of 30 mg/kg Nab-rapamycin significantly inhibited in vivo tumor daily for 15 days. The dosing volume was 2 ml/kg. Caliper growth for HCT-116 tumors, achieving a tumor growth inhi measurements of the longest perpendicular tumor diameters bition of 71% against the HCT-116 tumor xenograft were performed twice per week to estimate the tumor volume. 30 (p<0.0001 versus control, ANOVA: FIG. 6A). A -9.7% Animals were sacrificed when their tumor reached 2 cm or weight loss was observed in the mice from Nab-rapamycin at when the mice became moribund. As shown in FIG. 7, in all 40 mg/kg, which is similar to the -10.7% weight loss for the three treatment groups, Nab-rapamycin was highly effective control group (FIG. 6B). against multiple myeloma. 35 What is claimed is: Example 13 1. A method of treating cancer in a human individual, comprising administering to an individual an effective Cytotoxic Activity of Nab-rapamycin in amount of a composition comprising nanoparticles that com Combination with AbraxaneTM Against HT29 prise rapamycin or a derivative thereof and albumin, wherein (Human Colon Carcinoma) Tumor Xenograft 40 the rapamycin or derivative thereof is coated with albumin, wherein the amount of the rapamycin or derivative thereof in The following example is disclosed in U.S. Ser. No. the effective amount of the composition is in the range of 1 1/359,286, which was filed Feb. 21, 2006 (i.e., U.S. Pat. Pub. about 54 mg to about 180 mg, and wherein the average diam No. 2006/0263434, published Nov. 23, 2006). Nude mice eter of the nanoparticle in the capsule is no greater than about were implanted with 10 HT29 cells on their right flanks. 45 200 nm. Treatment was initiated once the tumors were palpable and 2. The method of claim 1, wherein the cancer is a plasma were greater than 100-200 mm. The mice were randomly cytoma. Sorted into 4 groups (n=8 per group). Group 1 received saline 3. The method of claim 1, wherein the cancer is selected 3 times weekly for 4 weeks, i.v.; Group 2 received Abrax from the group consisting of multiple myeloma, pancreatic aneTM at 10 mg/kg, daily for 5 days, i.p.; Group 3 received 50 cancer, brain cancer, prostate cancer, lung cancer, melanoma, Nab-rapamycin at 40 mg/kg, 3 times weekly for 4 weeks, i.v.; and breast cancer. and Group 4 received both Nab-rapamycin (40 mg/kg, 3 times 4. The method of claim 1, wherein the cancer is bladder weekly for 4 weeks, i.v.) and AbraxaneTM (10 mg/kg, daily for cancer, renal cell carcinoma, mesothelioma, or neuroendo 5 days, i.p.). As shown in FIG. 4, the tumor Suppression was crine cancer. greater for the AbraxaneTM plus Nab-rapamycin combination 55 5. The method of claim 4, wherein the cancer is bladder therapy than for either single therapy group. CaCC. 6. The method of claim 1, wherein the albumin is human Example 14 serum albumin. 7. A unit dosage form for treatment of cancer in a human Cytotoxic Activity of Nab-rapamycin Against HT29 60 individual comprising (a) nanoparticles that comprise rapa (Human Colon Carcinoma) Tumor Xenograft mycin or a derivative thereof and albumin, wherein the rapa mycin or derivative thereof is coated with albumin, wherein The antitumor activity of Nab-rapamycin was examined the amount of the rapamycin or derivative thereof in the unit using HT29 human colon carcinoma xenograft in mice. Male dosage form is in the range of about 54 mg to about 180 mg. athymic mice (3 per group) were implanted with 10 HT29 65 and (b) a pharmaceutically acceptable carrier, and wherein cells on their right flanks and allowed to grow to ~100 mm. the average diameter of the nanoparticles in the composition The mice were then intravenously administered with either is no greater than about 200 nm. US 8,911,786 B2 83 84 8. A kit comprising (a) nanoparticles that comprise rapa 19. The method of claim 10, wherein the chemotherapy mycin or a derivative thereof and albumin, wherein the rapa comprises administering to the individual a tyrosine kinase mycin or derivative thereof is coated with albumin, wherein inhibitor. 20. The method of claim 19, wherein the tyrosine kinase the amount of the rapamycin orderivative thereof in the kit is inhibitor is erlotinib. in the range of about 54 mg to about 180 mg, and (b) instruc 21. The method of claim 1, wherein the composition is tions for using the kit in treating cancer in a human individual, administered via an intravenous, intraarterial, intraperitoneal, wherein the average diameter of the nanoparticles in the com intravesicular, subcutaneous, or intrathecal route. position is no greater than about 200 nm. 22. The method of claim 9, wherein the composition is 9. A method of treating cancer in a human individual com administered via an intravenous, intraarterial, intraperitoneal, prising (a) a first therapy comprising administering to an 10 intravesicular, Subcutaneous, or intrathecal route. individual an effective amount of a composition comprising 23. The method of claim 1, wherein the composition com nanoparticles that comprise rapamycin or a derivative thereof prises nanoparticles comprising rapamycin. and albumin, where the rapamycin or derivative thereof is 24. The method of claim 23, wherein the composition coated with albumin and (b) a second therapy selected from comprises nanoparticles comprising rapamycin and human Second therapy selected from the group consisting of chemo 15 serum albumin. therapy, radiation therapy, surgery, hormone therapy, gene 25. The method of claim 9, wherein the composition com therapy, bone marrow transplantation, stem cell transplanta prises nanoparticles comprising rapamycin. tion, targeted therapy, cryotherapy, ultrasound therapy, and 26. The method of claim 25, wherein the composition immunotherapy, wherein the amount of the rapamycin or comprises nanoparticles comprising rapamycin and human derivative thereof in the effective amount of the composition serum albumin. is in the range of about 54 mg to about 180 mg, wherein the 27. The method of claim 23, wherein the cancer is bladder average diameter of the nanoparticles in the composition is no cancer, renal cell carcinoma, mesothelioma, or neuroendo greater than about 200 nm. crine cancer. 10. The method of claim 9, wherein the second therapy is 28. The method of claim 27, wherein the cancer is bladder chemotherapy. 25 CaCC. 11. The method of claim 10, wherein chemotherapy com 29. The method of claim 24, wherein the cancer is bladder prises administration of a chemotherapeutic selected from the cancer, renal cell carcinoma, mesothelioma, or neuroendo group consisting of antimetabolite agents, platinum-based crine cancer. 30. The method of claim 29, wherein the cancer is bladder agents, alkylating agents, tyrosine kinase inhibitors, anthra 30 cycline antibiotics, Vinca alkloids, proteasome inhibitors, and CaCC. topoisomerase inhibitors. 31. The method of claim 25, wherein the cancer is bladder 12. The method of claim 9, wherein the cancer is selected cancer, renal cell carcinoma, mesothelioma, or neuroendo from the group consisting of multiple myeloma, pancreatic crine cancer. cancer, prostate cancer, lung cancer, melanoma, and breast 32. The method of claim 31, wherein the cancer is bladder 35 CaCC. CaCC. 13. The method of claim 9, wherein the cancer is bladder 33. The method of claim 25, wherein the cancer is bladder cancer, renal cell carcinoma, mesothelioma, or neuroendo cancer, renal cell carcinoma, mesothelioma, or neuroendo crine cancer. crine cancer. 34. The method of claim 33, wherein the cancer is bladder 14. The method of claim9, wherein the first therapy and the 40 Second therapy are conducted sequentially. CaCC. 15. The method of claim 9, wherein the albumin is human 35. The method of claim 1, wherein the amount of the serum albumin. rapamycin orderivative thereof in the effective amount of the 16. The method of claim 10, wherein the chemotherapy composition is in the range of about 75 mg to about 100 mg. comprises administering to the individual an inhibitor of the 36. The method of claim 4, wherein the amount of the mTOR signaling pathway. 45 rapamycin orderivative thereof in the effective amount of the 17. The method of claim 10, wherein the chemotherapy composition is in the range of about 75 mg to about 100 mg. comprises administering to the individual an AKT kinase 37. The method of claim 23, wherein the amount of the inhibitor. rapamycin orderivative thereof in the effective amount of the 18. The method of claim 17, wherein AKT kinase inhibitor composition is in the range of about 75 mg to about 100 mg. is perifosine. ck ck ck ck ck