<<

US 2011 016O159A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2011/0160159 A1 Ryan (43) Pub. Date: Jun. 30, 2011

(54) TREATMENT OF 61/381.851, filed on Sep. 10, 2010, provisional appli cation No. 61/332,150, filed on May 6, 2010. (76) Inventor: John Ryan, Philadelphia, PA (US) Publication Classification 21) Appl. No.: 12/883,084 (51) Int. Cl. (21) Appl. No 9 A63L/724 (2006.01) 22) Filed: Sep.15, 2010 A6IP35/00 (2006.01) (22) File ep. 15, (52) U.S. Cl...... 514/58 Related U.S. Application Data (57) ABSTRACT (60) Provisional application No. 61/242,752, filed on Sep. Provided are methods relating to compositions that include a 15, 2009, provisional application No. 61/317,039, CDP-, e.g., a CDP- or filed on Mar. 24, 2010, provisional application No. camptothecin derivative conjugate, e.g., CRLX101.

Structure and description of CDP-camptothecin conjugate "CRLX101."

O ) Oi O O Y v/OH 4 HO O O HO O O HNS O OH O

N t H N-SS- HO -S o/). N-), O O OHIOO

O O YO O

In the above : n = about 77 or the molecular weight of the PEG moiety is from about 3060 to about 3740 (e.g., about 3400) Da; m at is from about 10 to about 18 (e.g., about 14), the molecular weight of the polymer backbone (i.e., the polymer minus the CPT-gly, which results in the Cysteine moieties having a free-C(O)CH) is from about 48 to about 85 kDa, the polydispersity of the polymer backbone is less than about 2.2, and the loading of the CPT onto the polymer backbone is from about 6 to about 13% by weight, wherein 13% is theoretical maximum, meaning, in some instances, one or more of the cysteine residues has a free-C(O)OH (i.e., it lacks the CPT-gly). Patent Application Publication Jun. 30, 2011 Sheet 1 of 4 US 2011/O16O159 A1

FIG. 1A FIG. 1 B

Patent Application Publication Jun. 30, 2011 Sheet 2 of 4 US 2011/O16O159 A1

F.G. 2A FIG. 2B

A B 30

es S. s So 220 X S c s c s 3 O 10 SS

0 25 50 75 100 125 150. 175 O 0-24 hrs 24-48 hrs Time (hr) Patent Application Publication Jun. 30, 2011 Sheet 3 of 4 US 2011/O16O159 A1

F.G. 3A FIG. 3B

250 ng total 500 ng total protein SC DNA Wock rx Pre Day 2 Day 25 Pe. Day 2 Day 25 relaxed

Relaxed Relaxed form form

Supercoiled DNA -->

F.G. 3C Patent Application Publication Jun. 30, 2011 Sheet 4 of 4 US 2011/O16O159 A1

Figure 4: Structure and description of CDP-camptothecin conjugate "CRLX101."

O O

OH O

O- O ()- O HHO O O O O O

in the above structure: n = about 77 or the molecular weight of the PEG moiety is from about 3060 to about 3740 (e.g., about 3400) Da; m F is from about 10 to about 18 (e.g., about 14); the molecular weight of the polymer backbone (i.e., the polymer minus the CPT-gly, which results in the cysteine moieties having a free -C(O)OH) is from about 48 to about 85 kDa, the polydispersity of the polymer backbone is less than about 2.2, and the loading of the CPT onto the polymer backbone is from about 6 to about 13% by weight, wherein 13% is theoretical maximum, meaning, in Some instances, one or more of the cysteine residues has a free -C(O)OH (i.e., it lacks the CPT-gly). US 2011/O160 159 A1 Jun. 30, 2011

TREATMENT OF CANCER 0011. In an embodiment, the is provided at 18-60 mg/m/month, e.g., 18-30 mg/m/month, 24-30 mg/m2/ CLAIM OF PRIORITY month or 36-60 mg/m/month. 0012. In an embodiment, the conjugate includes a topoi 0001. This application claims priority to U.S. Ser. No. somerase I inhibitor and/or a topoisomerase II inhibitor. In an 61/242,752, filed Sep. 15, 2009; U.S. Ser. No. 61/317,039, embodiment, the conjugate includes atopoisomerase I inhibi filed Mar. 24, 2010; U.S. Ser. No. 61/332,150, filed May 6, tor or combination of topoisomerase I inhibitors, e.g., camp 2010; and U.S. Ser. No. 61/381,851, filed Sep. 10, 2010, the tothecin, , SN-38, , lamellarin D and contents of each of which are incorporated herein by refer derivatives thereof. In an embodiment, the conjugate includes CCC. a topoisomerase II inhibitor or a combination of topoi Somerase II inhibitors, e.g., eptoposide, tenoposide, doxoru BACKGROUND OF THE INVENTION bicin and derivatives thereof. In one embodiment, the conju 0002 Drug delivery and dosing of small molecule thera gate includes a combination of one or more topoisomerase I peutic agents, such as camptothecin, can be problematic due inhibitors and one or more topoisomerase II inhibitors. In an to a number issues including half-life, toxicity, distribution embodiment, the CDP-topoisomerase inhibitor conjugate is a etc. CDP-camptothecin or camptothecin derivate conjugate, e.g., a CDP-camptothecin or camptothecin derivative conjugate SUMMARY OF THE INVENTION described herein, e.g., CRLX101. 0013. In one embodiment, the CDP-topoisomerase inhibi 0003. In one aspect, the disclosure features, a method of tor conjugate, particle or composition, e.g., a CDP-camptoth treating a proliferative disorder, e.g., a cancer, in a subject. ecin or camptothecin derivative, a CDP-camptothecin or The method comprises: camptothecin derivative conjugate, particle or composition 0004 providing an initial administration of a CDP-topoi described herein, e.g., CRLX101, is administered by intrave Somerase inhibitor conjugate, particle or composition, e.g., a nous administration over a period equal to or less than about CDP-camptothecin conjugate, particle or composition or 30 minutes, 45 minutes, 60 minutes, 90 minutes, 120 minutes, camptothecin derivative conjugate, particle or composition, 150 minutes, or 180 minutes. In one embodiment, the CDP e.g., a CDP-camptothecin conjugate, particle or composition topoisomerase inhibitor conjugate, particle or composition, or camptothecin derivative conjugate, particle or composition e.g., a CDP-camptothecin or camptothecin derivative conju described herein, e.g., CRLX101, to said subject at a dosage gate, particle or composition, e.g., the CDP-camptothecin or of 6 mg/m, 7 mg/m, 8 mg/m, 9 mg/m, 10 mg/m, 11 camptothecin derivative conjugate, particle or composition mg/m, 12 mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 described herein, e.g. CRLX101, is administered at a dosage mg/m, 17 mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 of 6 mg/m, 7 mg/m, 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 12 mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m. mg/m, 17 mg/m, 18 mg/m. 19 mg/m. 20 mg/m, 21 (wherein said dosage is expressed in mg of drug, as opposed mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 to mg of conjugate) and mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m by 0005 optionally, providing one or more subsequent intravenous administration over a period equal to or less than administrations of said CDP-topoisomerase inhibitor conju about 30 minutes, 45 minutes, 60 minutes or 90 minutes, e.g., gate, particle or composition, e.g., a CDP-camptothecin con a period equal to or less than 30 minutes, 45 minutes or 60 jugate, particle or composition or camptothecin derivative minutes. conjugate, particle or composition, e.g., a CDP-camptothecin 0014. In one embodiment, the CDP-topoisomerase inhibi conjugate, particle or composition or camptothecin derivative tor conjugate, particle or composition, e.g., a CDP-camptoth conjugate, particle or composition described herein, e.g., ecin or camptothecin derivative, a CDP-camptothecin or CRLX101, at a dosage of 6 mg/m, 7 mg/m, 8 mg/m, 9 camptothecin derivative conjugate, particle or composition mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 mg/m, 14 described herein, e.g., CRLX101, is administered by intrave mg/m, 15 mg/m, 16 mg/m, 17 mg/m, 18 mg/m, 19 nous administration overa period of about 12 hours, 15 hours, mg/m. 20 mg/m, 21 mg/m, 22 mg/m, 23 mg/m, 24 18 hours, 21 hours, 24 hours, 27 hours, or 30 hours. In one mg/m, 25 mg/m, 26 mg/m, 27 mg/m, 28 mg/m, 29 embodiment, the CDP-topoisomerase inhibitor conjugate, mg/m or 30 mg/m, wherein each subsequent administration particle or composition, e.g., a CDP-camptothecin or camp is provided, independently, between 9, 10, 11, 12, 13, 14, 15 tothecin derivative conjugate, particle or composition, e.g., or 16 days after the previous, e.g., the initial, administration, the CDP-camptothecin or camptothecin derivative conjugate, to thereby treat the proliferative disorder. particle or composition described herein, e.g. CRLX101, is 0006. In an embodiment, the dosage of at least 2, 3, 4, 5, 6, administered at a dosage of 6 mg/m, 7 mg/m, 8 mg/m, 9 7, 8, 9, 10, 12, 15 or 20 administrations is the same. mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 mg/m, 14 0007. In an embodiment, the time between at least 2, 3, 4, mg/m, 15 mg/m, 16 mg/m, 17 mg/m, 18 mg/m, 19 5, 6, 7, 8, 9, 10, 12, 15, or administrations is the same. mg/m. 20 mg/m, 21 mg/m, 22 mg/m, 23 mg/m, 24 0008. In an embodiment, each subsequent administration mg/m, 25 mg/m, 26 mg/m, 27 mg/m, 28 mg/m, 29 is administered 12-16, e.g., 14, days after the previous admin mg/m or 30 mg/m by intravenous administration over a istration. period of about 12 hours, 15 hours, 18 hours, 21 hours, 24 0009. In an embodiment, at least 2,3,4,5,6,7,8,9, 10, 12, hours, 27 hours, or 30 hours. Preferably, the CDP-topoi 15, 20, 50 or 100 administrations are administered to said Somerase inhibitor conjugate, particle or composition, e.g., a Subject. CDP-camptothecin or camptothecin derivative conjugate, 0010. In an embodiment, the drug is provided at 12-17 particle or composition, e.g., the CDP-camptothecin or camp mg/m/administration, e.g., 12-15 mg/m/administration. tothecin derivative conjugate, particle or composition US 2011/O160 159 A1 Jun. 30, 2011

described herein, e.g. CRLX101, is administered at a dosage resistant to a platinum based agent (e.g., , cispl of 15 mg/m, 16 mg/m, 17 mg/m, 18 mg/m. 19 mg/m. 20 atin, ) and/or a (e.g., , , mg/m, 21 mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 or ). mg/m, 26 mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 0020. In an embodiment, the method includes an initial mg/m by intravenous administration over a period of about administration of CRLX101 to said subject at a dosage of 12 12 hours, 15 hours, 18 hours, 21 hours, 24 hours, 27 hours, or mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 30 hours. mg/m, and 0015. In one embodiment, the CDP-topoisomerase inhibi 0021 one or more subsequent administrations of tor conjugate, particle or composition, e.g., a CDP-camptoth CRLX101 to said subject, at a dosage of 12 mg/m, 13 ecin or camptothecin derivative conjugate, particle or com mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 mg/m, e.g., at position, e.g., the CDP-camptothecin or camptothecin the same dosage as the initial dosage, wherein each Subse derivative conjugate, particle or composition described quent administration is administered, independently, 12-16, herein, e.g. CRLX101, is administered at a dosage of 6 e.g., 14, days after the previous, e.g., the initial, administra mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 tion, and the cancer is, e.g., ovarian cancer. In one embodi mg/m, 13 mg/m, or 14 mg/m twice a day, and optionally, ment, the ovarian cancer is refractory, relapsed or resistant to one or more Subsequent administrations of said CDP-topoi a platinum-based agent (e.g., carboplatin, , oxalipl Somerase inhibitor conjugate, particle or composition, e.g., a atin). In one embodiment, the CRLX101 is administered by CDP-camptothecin conjugate, particle or composition or intraperitoneal administration. camptothecin derivative conjugate, particle or composition, e.g., a CDP-camptothecin conjugate, particle or composition 0022. In an embodiment, the method includes an initial or camptothecin derivative conjugate, particle or composition administration of CRLX101 to said subject at a dosage of 16 described herein, e.g., CRLX101, is given at a dosage of 6 mg/m, 17 mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 13 mg/m, or 14 mg/m twice a day, wherein each mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, and Subsequent administration is provided, independently, 0023 one or more subsequent administrations of between 9, 10, 11, 12, 13, 14, 15 or 16 days after the previous, CRLX101 to said subject, at a dosage of 16 mg/m, 17 e.g., the initial, administration, to thereby treat the prolifera mg/m, 18 mg/m, 19 mg/m. 20 mg/m, 21 mg/m, 22 tive disorder. In one embodiment, the second daily dose is mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 given 4,5,6,7,8,9, 10, 12, 13, 14, 15, 16, 17, 18, 19, 20 hours mg/m, 28 mg/m, 29 mg/m or 30 mg/m, e.g., at the same after the initial daily dose. dosage as the initial dosage, wherein each subsequent admin 0016. In an embodiment, the method includes an initial istration is administered, independently, 12-16, e.g., 14, days administration of CRLX101 to said subject at a dosage of 12 after the previous, e.g., the initial, administration, and the mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 cancer is, e.g., ovarian cancer. In one embodiment, the ova mg/m and rian cancer is refractory, relapsed or resistant to a platinum 0017 one or more subsequent administrations of based agent (e.g., carboplatin, cisplatin, oxaliplatin). In one CRLX101 to said subject, at a dosage of 12 mg/m, 13 embodiment, the CRLX101 is administered by intraperito mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 mg/m, e.g., at neal administration. the same dosage as the initial dosage, wherein each Subse 0024. In an embodiment, the method includes an initial quent administration is administered, independently, 12-16, administration of CRLX101 to said subject at a dosage of 12 e.g., 14, days after the previous, e.g., the initial, administra mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 tion, and the cancer is, e.g., lung cancer, e.g., non-Small mg/m, and lung cancer and/or Small cell lung cancer (e.g., squamous cell 0025 one or more subsequent administrations of non-Small cell lung cancer or squamous cell Small cell lung CRLX101 to said subject, at a dosage of 12 mg/m, 13 cancer). In one embodiment, the lung cancer is refractory, mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 mg/m, e.g., at relapsed or resistant to a platinum based agent (e.g., carbopl the same dosage as the initial dosage, wherein each Subse atin, cisplatin, oxaliplatin) and/or a taxane (e.g., docetaxel, quent administration is administered, independently, 12-16, paclitaxel, larotaxel or cabazitaxel). e.g., 14, days after the previous, e.g., the initial, administra 0.018. In an embodiment, the method includes an initial tion, and the cancer is, e.g., gastric cancer, e.g., gastroesoph administration of CRLX101 to said subject at a dosage of 16 ageal, upper gastric or lower gastric cancer. mg/m, 17 mg/m, 18 mg/m. 19 mg/m. 20 mg/m, 21 0026. In an embodiment, the method includes an initial mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 administration of CRLX101 to said subject at a dosage of 16 mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, and mg/m, 17 mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 0019 one or more subsequent administrations of mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 CRLX101 to said subject, at a dosage of 16 mg/m, 17 mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, and mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 mg/m, 22 0027 one or more subsequent administrations of mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 CRLX101 to said subject, at a dosage of 16 mg/m, 17 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, e.g., at the same mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 mg/m, 22 dosage as the initial dosage, wherein each Subsequent admin mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 istration is administered, independently, 12-16, e.g., 14, days mg/m, 28 mg/m, 29 mg/m or 30 mg/m, e.g., at the same after the previous, e.g., the initial, administration, and the dosage as the initial dosage, wherein each Subsequent admin cancer is, e.g., lung cancer, e.g., non-Small cell lung cancer istration is administered, independently, 12-16, e.g., 14, days and/or small cell lung cancer (e.g., squamous cell non-Small after the previous, e.g., the initial, administration, and the cell lung cancer or squamous cell Small cell lung cancer). In cancer is, e.g., gastric cancer, e.g., gastroesophageal, upper one embodiment, the lung cancer is refractory, relapsed or gastric or lower gastric cancer. US 2011/O160 159 A1 Jun. 30, 2011

0028. In an embodiment, the method includes an initial 0038. In an embodiment, the method includes an initial administration of CRLX101 to said subject at a dosage of 12 administration of CRLX101 to said subject at a dosage of 16 mg/m, 13 mg/m, 14 mg/m or 15 mg/m, and mg/m, 17 mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 0029 one or more subsequent administrations of mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 CRLX101 to said subject, at a dosage of 12 mg/m, 13 mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, and mg/m, 14 mg/m or 15 mg/m, e.g., at the same dosage as the initial dosage, wherein each Subsequent administration is 0039 one or more subsequent administrations of administered, independently, 12-16, e.g., 14, days after the CRLX101 to said subject, at a dosage of 16 mg/m, 17 previous, e.g., the initial, administration, and the cancer is, mg/m, 18 mg/m, 19 mg/m. 20 mg/m, 21 mg/m, 22 e.g., pancreatic cancer. mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 0030. In an embodiment, the method includes an initial mg/m, 28 mg/m, 29 mg/m or 30 mg/m, e.g., at the same administration of CRLX101 to said subject at a dosage of 16 dosage as the initial dosage, wherein each Subsequent admin mg/m, 17 mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 istration is administered, independently, 12-16, e.g., 14, days mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 after the previous, e.g., the initial, administration, and the mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, and cancer is, e.g., breast cancer, e.g., estrogen receptor positive 0031 one or more subsequent administrations of breast cancer, estrogen receptor negative breast cancer, CRLX101 to said subject, at a dosage of 16 mg/m, 17 HER-2 positive breast cancer, HER-2 negative breast cancer, mg/m, 18 mg/m, 19 mg/m, 20 mg/m, 21 mg/m, 22 triple negative breast cancer or inflammatory breast cancer. mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 0040. In an embodiment, the cancer is a cancer described mg/m, 28 mg/m, 29 mg/m or 30 mg/m, e.g., at the same herein. For example, the cancer can be a cancer of the bladder dosage as the initial dosage, wherein each Subsequent admin (including accelerated and metastatic bladder cancer), breast istration is administered, independently, 12-16, e.g., 14, days (e.g., estrogen receptor positive breast cancer, estrogen recep after the previous, e.g., the initial, administration, and the tor negative breast cancer, HER-2 positive breast cancer, cancer is, e.g., pancreatic cancer. HER-2 negative breast cancer, triple negative breast cancer, 0032. In an embodiment, the method includes an initial inflammatory breast cancer), colon (including colorectal can administration of CRLX101 to said subject at a dosage of 12 cer), kidney, liver, lung (including Small cell lung cancer and mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 non-Small cell lung cancer (including adenocarcinoma, squa mg/m, and mous cell carcinoma, bronchoalveolar carcinoma and large 0033 one or more subsequent administrations of cell carcinoma)), genitourinary tract, e.g., ovary (including CRLX101 to said subject, at a dosage of 12 mg/m, 13 fallopian, endometrial and peritoneal ), cervix, pros mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 mg/m, e.g., at tate and testes, lymphatic system, rectum, larynx, pancreas the same dosage as the initial dosage, wherein each Subse (including exocrine pancreatic carcinoma), stomach (e.g., quent administration is administered, independently, 12-16, gastroesophageal, upper gastric or lower gastric cancer), gas e.g., 14, days after the previous, e.g., the initial, administra trointestinal cancer (e.g., anal cancer), gallbladder, thyroid, tion, and the cancer is, e.g., . lymphoma (e.g., Burkitt's, Hodgkin's or non-Hodgkin's lym 0034. In an embodiment, the method includes an initial phoma), (e.g., acute myeloid leukemia), Ewing's administration of CRLX101 to said subject at a dosage of 16 sarcoma, nasoesophageal cancer, nasopharyngeal cancer, mg/m, 17 mg/m, 18 mg/m. 19 mg/m. 20 mg/m, 21 neural and glial cell cancers (e.g., glioblastoma multiforme), mg/m, 22 mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 and head and neck. Preferred cancers include breast cancer mg/m, 27 mg/m, 28 mg/m, 29 mg/m or 30 mg/m, and (e.g., metastatic or locally advanced breast cancer), prostate 0035 one or more subsequent administrations of cancer (e.g., hormone refractory prostate cancer), renal cell CRLX101 to said subject, at a dosage of 16 mg/m, 17 carcinoma, lung cancer (e.g., Small cell lung cancer and non mg/m, 18 mg/m, 19 mg/m. 20 mg/m, 21 mg/m, 22 Small cell lung cancer (including adenocarcinoma, squamous mg/m, 23 mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 cell carcinoma, bronchoalveolar carcinoma and large cell mg/m, 28 mg/m, 29 mg/m or 30 mg/m, e.g., at the same carcinoma)), pancreatic cancer, gastric cancer (e.g., gastroe dosage as the initial dosage, wherein each Subsequent admin sophageal, upper gastric or lower gastric cancer), colorectal istration is administered, independently, 12-16, e.g., 14, days cancer, squamous cell cancer of the head and neck, ovarian after the previous, e.g., the initial, administration, and the cancer (e.g., advanced ovarian cancer, platinum-based agent cancer is, e.g., colorectal cancer. resistant or relapsed ovarian cancer), lymphoma (e.g., Bur 0036. In an embodiment, the method includes an initial kitt's, Hodgkin's or non-Hodgkin's lymphoma), leukemia administration of CRLX101 to said subject at a dosage of 12 (e.g., acute myeloid leukemia) and gastrointestinal cancer. mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 0041. In an embodiment, the cancer is ovarian cancer, mg/m, and colorectal, breast, lung, lymphoma or gastric cancer. In an 0037 one or more subsequent administrations of embodiment, the cancer is a cancer other than pancreatic CRLX101 to said subject, at a dosage of 12 mg/m, 13 cancer, renal cell carcinoma and/or lung cancer (e.g., non mg/m, 14 mg/m, 15 mg/m, 16 mg/m, or 17 mg/m, e.g., at Small cell lung cancer and/or Small cell lung cancer). In an the same dosage as the initial dosage, wherein each Subse embodiment, the cancer is a cancer other than pancreatic quent administration is administered, independently, 12-16, cancer, renal cell carcinoma, lung cancer (e.g., non-small cell e.g., 14, days after the previous, e.g., the initial, administra lung cancer and/or Small cell lung cancer) and/or ovarian tion, and the cancer is, e.g., breast cancer, e.g., estrogen CaCC. receptor positive breast cancer, estrogen receptor negative 0042. In one embodiment, the subject has not been admin breast cancer, HER-2 positive breast cancer, HER-2 negative istered a CDP-topoisomerase inhibitor conjugate, particle or breast cancer, triple negative breast cancer or inflammatory composition, e.g., a CDP-camptothecin or camptothecin breast cancer. derivative conjugate, particle or composition, a CDP-camp US 2011/O160 159 A1 Jun. 30, 2011

tothecin or camptothecin derivative conjugate, particle or 26, 27, 28, 29, 30 or 31 days, e.g., 28 days. In one embodi composition described herein, e.g., CRLX101, prior to the ment, when the CDP-topoisomerase inhibitor conjugate, par initial administration. ticle or composition is administered in combination with 0043. In an embodiment, the CDP-topoisomerase inhibi (e.g., liposomal doxorubicin), the dose at which tor conjugate, particle or composition is administered as a the CDP-topoisomerase inhibitor conjugate, particle or com first line treatment for the cancer. position is administered is 1%, 3%. 5%, 10%, 15%, 20%, 0044. In an embodiment, the CDP-topoisomerase inhibi 25%, 30% less than a dose described herein. tor conjugate, particle or composition is administered as a 0046. In an embodiment, the cancer has been sensitized to second, third or fourth line treatment for the cancer. In an atopoisomerase inhibitor, e.g., the Subject has received radia embodiment, the cancer is sensitive to one or more chemo tion and/or the subject has received a phosphatase inhibitor therapeutic agents, e.g., a platinum based agent, a taxane, an (e.g., okadiac acid) prior to the administration of the CDP alkylating agent, an (e.g., doxorubicin (e.g., topoisomerase inhibitor conjugate, particle or composition. liposomal doxorubicin)), an and/or a Vinca In one embodiment, the cancer is sensitized to topoisomerase alkaloid. In an embodiment, the cancer is a refractory, inhibitors, e.g., the Subject receives radiation in combination relapsed or resistant to one or more chemotherapeutic agents, with the administration of the CDP-topoisomerase inhibitor e.g., a platinum based agent, a taxane, an alkylating agent, an conjugate, particle or composition and/or the Subject is antimetabolite and/or a Vinca alkaloid. In one embodiment, administered a phosphatase inhibitor (e.g., okadiac acid) in the cancer is, e.g., ovarian cancer, and the ovarian cancer is combination with the administration of the CDP-topoi refractory, relapsed or resistant to a platinum based agent Somerase inhibitor conjugate, particle or composition. In one (e.g., carboplatin, cisplatin, oxaliplatin), a taxane (e.g., pacli embodiment, the cancer is sensitized or has been sensitized to taxel, docetaxel, larotaxel, cabazitaxel) and/or an anthracy topoisomerase inhibitors and the cancer is a glial cell cancer cline (e.g., doxorubicin (e.g., liposomal doxorubicin)). In one (e.g., glioblastoma multiforme) or head and neck cancer. embodiment, the cancer is, e.g., colorectal cancer, and the 0047. In one embodiment, the conjugate, particle or com cancer is refractory, relapsed or resistant to an antimetabolite position is administered in combination with one or more (e.g., an (e.g., , , ) additional chemotherapeutic agent, e.g., a chemotherapeutic and a analogue (e.g., , cytrarabine, agent (such as an angiogenesis inhibitor) or combination of , 5FU)) and/or a platinum based agent (e.g., car chemotherapeutic agents described herein. In one embodi boplatin, cisplatin, oxaliplatin). In one embodiment, the can ment, the conjugate, particle or composition is administered cer is, e.g., lung cancer, and the cancer is refractory, relapsed in combination with one or more of: a platinum-based agent or resistant to a taxane (e.g., paclitaxel, docetaxel, larotaxel, (e.g., carboplatin, cisplatin, oxaliplatin), a taxane (e.g., pacli cabazitaxel), a platinum based agent (e.g., carboplatin, cispl taxel, docetaxel, larotaxel, cabazitaxel), a Vinca alkaloid (e.g., atin, oxaliplatin), a Vinca alkaloid (e.g., , Vincris vinblastine, , , ), an antime tine, Vindesine, Vinorelbine), a vascular endothelial growth tabolite (e.g., an antifolate (e.g., pemetrexed, floXuridine, factor (VEGF) pathway inhibitor, an epidermal growth factor raltitrexed) and a (e.g., 5FU, capecitab (EGF) pathway inhibitor and/or an antimetabolite (e.g., an ine, cytrarabine, gemcitabine)), an alkylating agent (e.g., antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a , decarbazine, , , pyrimidine analogue (e.g., capecitabine, cytrarabine, gemcit ), a vascular endothelial growth factor (VEGF) abine, 5FU)). In one embodiment, the cancer is, e.g., breast pathway inhibitor, a poly ADP-ribose polymerase (PARP) cancer, and the cancer is refractory, relapsed or resistant to a inhibitor and an mTOR inhibitor. In one embodiment, when taxane (e.g., paclitaxel, docetaxel, larotaxel, cabazitaxel), a the CDP-topoisomerase inhibitor conjugate, particle or com vascular endothelial growth factor (VEGF) pathway inhibi position is administered in combination with an additional tor, an anthracycline (e.g., , doxorubicin (e.g., chemotherapeutic agent, the dose at which the CDP-topoi liposomal doxorubicin), , , ), a Somerase inhibitor conjugate, particle or composition is platinum-based agent (e.g., carboplatin, cisplatin, oxalipl administered is 1%, 3%, 5%, 10%, 15%, 20%, 25%, 30% less atin), and/or an antimetabolite (e.g., an antifolate (e.g., pem than a dose described herein. etrexed, floXuridine, raltitrexed) and a pyrimidine analogue 0048. In an embodiment, the method further comprises (e.g., capecitabine, cytrarabine, gemcitabine, 5FU)). In one administering to said subject a treatment that reduces one or embodiment, the cancer is, e.g., gastric cancer, and the cancer more side effect associated with administration of a CDP is refractory, relapsed or resistant to an antimetabolite (e.g., camptothecin or camptothecin derivative conjugate, particle an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a or composition, e.g., a treatment described herein. pyrimidine analogue (e.g., capecitabine, cytrarabine, gemcit 0049. In one aspect, the disclosure features, a method of abine, 5FU)) and/or a platinum-based agent (e.g., carbopl treating a proliferative disorder, e.g., a cancer, in a Subject, atin, cisplatin, oxaliplatin). e.g., a human Subject. The method comprises: 0045. In one embodiment, the subject has ovarian cancer 0050 providing an initial administration of a CDP-topoi that is refractory, relapsed or resistant to a platinum-based Somerase inhibitor conjugate, particle or composition, e.g., a agent, and the Subject is administered a CDP-topoisomerase CDP-camptothecin or camptothecin derivative conjugate, inhibitor conjugate, particle or composition, e.g., a CDP particle or composition, e.g., a CDP-camptothecin or camp topoisomerase inhibitor conjugate, particle or composition tothecin derivative conjugate, particle or composition described herein. In one embodiment, the CDP-topoi described herein, e.g., CRLX101, to said Subject at a dosage Somerase inhibitor conjugate, particle or composition is of 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 mg/m, 14 administered in combination with doxorubicin (e.g., liposo mg/m, 15 mg/m, 16 mg/m, 17 mg/m, 18 mg/m, 19 mal doxorubicin). In one embodiment, the doxorubicin (e.g., mg/m. 20 mg/m, 21 mg/m, 22 mg/m, 23 mg/m, 24 the liposomal doxorubicin) is administered at a dose of about mg/m, 25 mg/m, 26 mg/m, 27 mg/m, 28 mg/m, 29 20 mg/m, about 30 mg/m or about 40 mg/m, every 24, 25, mg/m, 30 mg/m, 31 mg/m, 32 mg/m, 33 mg/m, 34 US 2011/O160 159 A1 Jun. 30, 2011 mg/m, 35 mg/m or 36 mg/m (wherein said dosage is mg/m, 15 mg/m, 16 mg/m, 17 mg/m, 18 mg/m, 19 expressed in mg of drug, as opposed to mg of conjugate) and mg/m. 20 mg/m, 21 mg/m, 22 mg/m, 23 mg/m, 24 0051 optionally, providing one or more subsequent mg/m, 25 mg/m, 26 mg/m, 27 mg/m, 28 mg/m, 29 administrations of said CDP-topoisomerase inhibitor conju mg/m, 30 mg/m, 31 mg/m, 32 mg/m, 33 mg/m, 34 gate, particle or composition, e.g., a CDP-camptothecin or mg/m, 35 mg/m or 36 mg/m by intravenous administration camptothecin derivative conjugate, particle or composition, over a period equal to or less than about 30 minutes, 45 e.g., a CDP-camptothecin or camptothecin derivative conju minutes, 60 minutes or 90 minutes, e.g., a period equal to or gate, particle or composition described herein, e.g., less than 30 minutes, 45 minutes or 60 minutes. CRLX101, at a dosage of 6 mg/m, 7 mg/m, 8 mg/m, 9 0059. In one embodiment, the CDP-topoisomerase inhibi mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 mg/m, 14 tor conjugate, particle or composition, e.g., a CDP-camptoth mg/m, 15 mg/m, 16 mg/m, 17 mg/m, 18 mg/m. 19 ecin or camptothecin derivative, a CDP-camptothecin or mg/m. 20 mg/m, 21 mg/m, 22 mg/m, 23 mg/m, 24 camptothecin derivative conjugate, particle or composition mg/m, 25 mg/m, 26 mg/m, 27 mg/m, 28 mg/m, 29 described herein, e.g., CRLX101, is administered by intrave mg/m, 30 mg/m, 31 mg/m, 32 mg/m, 33 mg/m, 34 nous administration overa period of about 12 hours, 15 hours, mg/m, 35 mg/m or 36 mg/m, wherein each subsequent 18 hours, 21 hours, 24 hours, 27 hours or 30 hours. In one administration is provided, independently, between 17, 18, embodiment, the CDP-topoisomerase inhibitor conjugate, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or 31 days after particle or composition, e.g., a CDP-camptothecin or camp the previous, e.g., the initial, administration, to thereby treat tothecin derivative conjugate, particle or composition, e.g., the proliferative disorder. the CDP-camptothecin or camptothecin derivative conjugate, 0052. In an embodiment, the dosage of at least 2, 3, 4, 5, 6, particle or composition described herein, e.g. CRLX101, is 7, 8, 9, 10, 12, 15 or 20 administrations are the same. administered at a dosage of 15 mg/m, 16 mg/m, 17 mg/m. 0053. In an embodiment, the time between at least 2, 3, 4, 18 mg/m, 19 mg/m, 20 mg/m, 21 mg/m, 22 mg/m, 23 5, 6, 7, 8, 9, 10, 12 15, or administrations is the same. mg/m, 24 mg/m, 25 mg/m, 26 mg/m, 27 mg/m, 28 0054. In an embodiment, each subsequent administration mg/m, 29 mg/m, 30 mg/m, 31 mg/m, 32 mg/m, 33 is administered 19-23, e.g., 21, or 25-29, e.g., 27 or 28 days mg/m, 34 mg/m, 35 mg/m or 36 mg/m by intravenous after the previous administration. administration over a period of about 12 hours, 15 hours, 18 0055. In an embodiment, at least 2,3,4,5,6,7,8,9, 10, 12, hours, 21 hours, 24 hours, 27 hours or 30 hours. 15, 20, 50 or 100 administrations are administered to said 0060. In one embodiment, the CDP-topoisomerase inhibi subject. tor conjugate, particle or composition, e.g., a CDP-camptoth 0056. In an embodiment, the drug is provided at 18-60 ecin or camptothecin derivative conjugate, particle or com mg/m/month, e.g., 18-30 mg/m/month or 36-60 mg/m/ position, e.g., the CDP-camptothecin or camptothecin month. In one embodiment, when the drug is provided in derivative conjugate, particle or composition described combination with one or more additional chemotherapeutic herein, e.g. CRLX101, is administered at a dosage of 6 agent, e.g., a chemotherapeutic agent described herein, the mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 drug is provided at 6-12 mg/m/month. mg/m, 13 mg/m, or 14 mg/m twice a day, and optionally, 0057. In an embodiment, the conjugate includes a topoi one or more Subsequent administrations of said CDP-topoi somerase I inhibitor and/or a topoisomerase II inhibitor. In an Somerase inhibitor conjugate, particle or composition, e.g., a embodiment, the conjugate includes atopoisomerase I inhibi CDP-camptothecin conjugate, particle or composition or tor or combination of topoisomerase I inhibitors, e.g., camp camptothecin derivative conjugate, particle or composition, tothecin, irinotecan, SN-38, topotecan, lamellarin D and e.g., a CDP-camptothecin conjugate, particle or composition derivatives thereof. In an embodiment, the conjugate includes or camptothecin derivative conjugate, particle or composition a topoisomerase II inhibitor or a combination of topoi described herein, e.g., CRLX101, is given at a dosage of 6 Somerase II inhibitors, e.g., , tenoposide, doxorubi mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 cin and derivatives thereof. In one embodiment, the conjugate mg/m, 13 mg/m, or 14 mg/m twice a day, wherein each includes a combination of one or more topoisomerase I Subsequent administration is provided, independently, inhibitors and one or more topoisomerase II inhibitors. In an between 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 20 embodiment, the CDP-topoisomerase inhibitor conjugate is a or 31 days after the previous, e.g., the initial, administration, CDP-camptothecin or camptothecin derivate conjugate, e.g., to thereby treat the proliferative disorder. In one embodiment, a CDP-camptothecin or camptothecin derivative conjugate the second daily dose is given 4, 5, 6,7,8,9, 10, 12, 13, 14, 15, described herein, e.g., CRLX101. 16, 17, 18, 19, 20 hours after the initial daily dose. 0058. In one embodiment, the CDP-topoisomerase inhibi 0061. In an embodiment, the method includes an initial tor conjugate, particle or composition, e.g., a CDP-camptoth administration of CRLX101 to said subject at a dosage of 6 ecin or camptothecin derivative, a CDP-camptothecin or mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 camptothecin derivative conjugate, particle or composition mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 described herein, e.g., CRLX101, is administered by intrave mg/m or 18 mg/m, and nous administration over a period equal to or less than about 0062 one or more subsequent administrations of 30 minutes, 45 minutes, 60 minutes, 90 minutes, 120 minutes, CRLX101 to said subject, at a dosage of 6 mg/m, 7 mg/m. 150 minutes, or 180 minutes. In one embodiment, the CDP 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 topoisomerase inhibitor conjugate, particle or composition, mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m or 18 e.g., a CDP-camptothecin or camptothecin derivative conju mg/m, e.g., at the same dosage as the initial dosage, wherein gate, particle or composition, e.g., the CDP-camptothecin or each Subsequent administration is administered, indepen camptothecin derivative conjugate, particle or composition dently, 19-22, e.g., 21 days after the previous, e.g., the initial, described herein, e.g. CRLX101, is administered at a dosage administration, and the cancer is, e.g., lung cancer, e.g., non of 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 mg/m, 14 Small cell lung cancer and/or Small cell lung cancer (e.g., US 2011/O160 159 A1 Jun. 30, 2011 squamous cell non-small cell lung cancer or squamous cell mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 Small cell lung cancer). In one embodiment, the lung cancer is mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 refractory, relapsed or resistant to a platinum based agent mg/m or 18 mg/m, and (e.g., carboplatin, cisplatin, oxaliplatin) and/or a taxane (doc 0072 one or more subsequent administrations of etaxel, paclitaxel, larotaxel or cabazitaxel). CRLX101 to said subject, at a dosage of 6 mg/m, 7 mg/m. 0063. In an embodiment, the method includes an initial 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 administration of CRLX101 to said subject at a dosage of 6 mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m or 18 mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m, e.g., at the same dosage as the initial dosage, wherein mg/m or 18 mg/m, and each Subsequent administration is administered, indepen 0064 one or more subsequent administrations of dently, 19-22, e.g., 21 days after the previous, e.g., the initial, CRLX101 to said subject, at a dosage of 6 mg/m, 7 mg/m. administration, and the cancer is, e.g., breast cancer, e.g., 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 estrogen receptor positive breast cancer, estrogen receptor mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m or 18 negative breast cancer, HER-2 positive breast cancer, HER-2 mg/m, e.g., at the same dosage as the initial dosage, wherein negative breast cancer, triple negative breast cancer or inflam each Subsequent administration is administered, indepen matory breast cancer. dently, 19-22, e.g., 21, days after the previous, e.g., the initial, 0073. In an embodiment, the cancer is a cancer described administration, and the cancer is, e.g., ovarian cancer. In one herein. For example, the cancer can be a cancer of the bladder embodiment, the ovarian cancer is refractory, relapsed or (including accelerated and metastatic bladder cancer), breast resistant to a platinum-based agent (e.g., carboplatin, cispl (e.g., estrogen receptor positive breast cancer, estrogen recep atin, oxaliplatin). In one embodiment, the CRLX101 is tor negative breast cancer, HER-2 positive breast cancer, administered by intraperitoneal administration. HER-2 negative breast cancer, triple negative breast cancer, 0065. In an embodiment, the method includes an initial inflammatory breast cancer), colon (including colorectal can administration of CRLX101 to said subject at a dosage of 6 cer), kidney, liver, lung (including Small cell lung cancer and mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 non-Small cell lung cancer (including adenocarcinoma, squa mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mous cell carcinoma, bronchoalveolar carcinoma and large mg/m or 18 mg/m, and cell carcinoma)), genitourinary tract, e.g., ovary (including 0066 one or more subsequent administrations of fallopian, endometrial and peritoneal cancers), cervix, pros CRLX101 to said subject, at a dosage of 6 mg/m, 7 mg/m. tate and testes, lymphatic system, rectum, larynx, pancreas 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 (including exocrine pancreatic carcinoma), stomach (e.g., mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m or 18 gastroesophageal, upper gastric or lower gastric cancer), gas mg/m, e.g., at the same dosage as the initial dosage, wherein trointestinal cancer (e.g., anal cancer), gallbladder, thyroid, each Subsequent administration is administered, indepen lymphoma (e.g., Burkitt's, Hodgkin's or non-Hodgkin's lym dently, 19-22, e.g., 21, days after the previous, e.g., the initial, phoma), leukemia (e.g., acute myeloid leukemia), Ewing's administration, and the cancer is, e.g., gastric cancer, e.g., sarcoma, nasoesophageal cancer, nasopharyngeal cancer, gastroesophageal, upper gastric or lower gastric cancer. neural and glial cell cancers (e.g., glioblastoma multiforme), 0067. In an embodiment, the method includes an initial and head and neck. Preferred cancers include breast cancer administration of CRLX101 to said subject at a dosage of 6 (e.g., metastatic or locally advanced breast cancer), prostate mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 cancer (e.g., hormone refractory prostate cancer), renal cell mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 carcinoma, lung cancer (e.g., Small cell lung cancer and non mg/m or 18 mg/m, and Small cell lung cancer (including adenocarcinoma, squamous 0068 one or more subsequent administrations of cell carcinoma, bronchoalveolar carcinoma and large cell CRLX101 to said subject, at a dosage of 6 mg/m, 7 mg/m. carcinoma)), pancreatic cancer, gastric cancer (e.g., gastroe 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 sophageal, upper gastric or lower gastric cancer), colorectal mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m or 18 cancer, squamous cell cancer of the head and neck, ovarian mg/m, e.g., at the same dosage as the initial dosage, wherein cancer (e.g., advanced ovarian cancer, platinum-based agent each Subsequent administration is administered, indepen resistant or relapsed ovarian cancer), lymphoma (e.g., Bur dently, 19-22, e.g., 21, days after the previous, e.g., the initial, kitt's, Hodgkin's or non-Hodgkin's lymphoma), leukemia administration, and the cancer is, e.g., pancreatic cancer. (e.g., acute myeloid leukemia) and gastrointestinal cancer. 0069. In an embodiment, the method includes an initial 0074. In an embodiment, the cancer is ovarian cancer, administration of CRLX101 to said subject at a dosage of 6 colorectal, breast, lung, lymphoma or gastric cancer. In an mg/m, 7 mg/m.8 mg/m.9 mg/m, 10 mg/m, 11 mg/m, 12 embodiment, the cancer is a cancer other than pancreatic mg/m, 13 mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 cancer, renal cell carcinoma and/or lung cancer (e.g., non mg/m or 18 mg/m, and Small cell lung cancer). In an embodiment, the cancer is a 0070 one or more subsequent administrations of cancer other than pancreatic cancer, renal cell carcinoma, CRLX101 to said subject, at a dosage of 6 mg/m, 7 mg/m. lung cancer (e.g., non-Small cell lung cancer) and/or ovarian 8 mg/m, 9 mg/m, 10 mg/m, 11 mg/m, 12 mg/m, 13 CaCC. mg/m, 14 mg/m, 15 mg/m, 16 mg/m, 17 mg/m or 18 0075. In one embodiment, the subject has not been admin mg/m, e.g., at the same dosage as the initial dosage, wherein istered a CDP-topoisomerase inhibitor conjugate, particle or each Subsequent administration is administered, indepen composition, e.g., a CDP-camptothecin or camptothecin dently, 19-22, e.g., 21, days after the previous, e.g., the initial, derivative conjugate, particle or composition, a CDP-camp administration, and the cancer is, e.g., colorectal cancer. tothecin or camptothecin derivative conjugate, particle or 0071. In an embodiment, the method includes an initial composition described herein, e.g., CRLX101, prior to the administration of CRLX101 to said subject at a dosage of 6 initial administration. US 2011/O160 159 A1 Jun. 30, 2011

0076. In an embodiment, the CDP-topoisomerase inhibi composition, e.g., a CDP-topoisomerase inhibitor conjugate, tor conjugate, particle or composition is administered as a particle or composition described herein is administered at a first line treatment for the cancer. dose and/or dosing regimen described herein and the doxo 0077. In an embodiment, the CDP-topoisomerase inhibi rubicin (e.g., the liposomal doxorubicin) is administered at a tor conjugate, particle or composition is administered as a dose of about 20 mg/m, about 30 mg/m or about 40 mg/m. second, third or fourth line treatment for the cancer. In an every 24, 25, 26, 27, 28, 29.30 or 31 days, e.g., 28 days. In one embodiment, the cancer is sensitive to one or more chemo embodiment, when the CDP-topoisomerase inhibitor conju therapeutic agents, e.g., a platinum-based agent, a taxane, an gate, particle or composition is administered in combination alkylating agent, an antimetabolite and/or a Vinca alkaloid. In with doxorubicin (e.g., liposomal doxorubicin), the dose at an embodiment, the cancer is a refractory, relapsed or resis which the CDP-topoisomerase inhibitor conjugate, particle tant to or composition is administered is 1%, 3%. 5%, 10%, 15%, 0078 one or more chemotherapeutic agents, e.g., a plati 20%, 25%, 30% less than a dose described herein. num-based agent, a taxane, an alkylating agent, an anthracy 0080. In an embodiment, the cancer has been sensitized to cline (e.g., doxorubicin (e.g., liposomal doxorubicin)), an topoisomerase inhibitors, e.g., the Subject has received radia antimetabolite and/or a Vinca alkaloid. In one embodiment, tion and/or the subject has received a phosphatase inhibitor the cancer is, e.g., ovarian cancer, and the ovarian cancer is (e.g., okadiac acid) prior to the administration of the CDP refractory, relapsed or resistant to a platinum-based agent topoisomerase inhibitor conjugate, particle or composition. (e.g., carboplatin, cisplatin, oxaliplatin), a taxane (e.g., pacli In an embodiment, the cancer is sensitized to topoisomerase taxel, docetaxel, larotaxel, cabazitaxel) and/or an anthracy inhibitors, e.g., the Subject receives radiation in combination cline (e.g., doxorubicin (e.g., liposomal doxorubicin)). In one with the administration of the CDP-topoisomerase inhibitor embodiment, the cancer is, e.g., colorectal cancer, and the conjugate, particle or composition and/or the Subject is cancer is refractory, relapsed or resistant to an antimetabolite administered a phosphatase inhibitor (e.g., okadiac acid) in (e.g., an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) combination with the administration of the CDP-topoi and a pyrimidine analogue (e.g., capecitabine, cytrarabine, Somerase inhibitor conjugate, particle or composition. In one gemcitabine, 5FU)) and/or a platinum-based agent (e.g., car embodiment, the cancer is sensitized or has been sensitized to boplatin, cisplatin, oxaliplatin). In one embodiment, the can topoisomerase inhibitors and the cancer is a glial cell cancer cer is, e.g., lung cancer, and the cancer is refractory, relapsed (e.g., glioblastoma multiforme) or head and neck cancer. or resistant to a taxane (e.g., paclitaxel, docetaxel, larotaxel, I0081. In one embodiment, the conjugate, particle or com cabaZitaxel), a platinum-based agent (e.g., carboplatin, cispl position is administered in combination with one or more atin, oxaliplatin), a Vinca alkaloid (e.g., vinblastine, Vincris additional chemotherapeutic agent, e.g., a chemotherapeutic tine, Vindesine, Vinorelbine), a vascular endothelial growth agent (such as an angiogenesis inhibitor) or combination of factor (VEGF) pathway inhibitor, an epidermal growth factor chemotherapeutic agents described herein. In one embodi (EGF) pathway inhibitor) and/or an antimetabolite (e.g., an ment, the conjugate, particle or composition is administered antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a in combination with one or more of a platinum based agent pyrimidine analogue (e.g., capecitabine, cytrarabine, gemcit (e.g., carboplatin, cisplatin, oxaliplatin), a taxane (e.g., pacli abine, 5FU)). In one embodiment, the cancer is, e.g., breast taxel, docetaxel, larotaxel, cabazitaxel), a Vinca alkaloid (e.g., cancer, and the cancer is refractory, relapsed or resistant to a vinblastine, Vincristine, Vindesine, Vinorelbine), an antime taxane (e.g., paclitaxel, docetaxel, larotaxel, cabazitaxel), a tabolite (e.g., an antifolate (e.g., floXuridine, premetrexed), a vascular endothelial growth factor (VEGF) pathway inhibi pyrimidine analogue (e.g., 5FU, capecitabine)), an alkylating tor, an anthracycline (e.g., daunorubicin, doxorubicin (e.g., agent (e.g., cyclophosphamide, decarbazine, melphalan, ifos liposomal doxorubicin), epirubicin, valrubicin, idarubicin), a famide, temozolomide), a vascular endothelial growth factor platinum-based agent (e.g., carboplatin, cisplatin, oxalipl (VEGF) pathway inhibitor, a poly ADP-ribose polymerase atin), and/or an antimetabolite (e.g., an antifolate (e.g., pem (PARP) inhibitor and an mTOR inhibitor. In one embodi etrexed, floXuridine, raltitrexed) and a pyrimidine analogue ment, when the CDP-topoisomerase inhibitor conjugate, par (e.g., capecitabine, cytrarabine, gemcitabine, 5FU)). In one ticle or composition is administered in combination with an embodiment, the cancer is, e.g., gastric cancer, and the cancer additional chemotherapeutic agent, the dose at which the is refractory, relapsed or resistant to an antimetabolite (e.g., CDP-topoisomerase inhibitor conjugate, particle or compo an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a sition is administered is 1%, 3%. 5%, 10%, 15%, 20%, 25%, pyrimidine analogue (e.g., capecitabine, cytrarabine, gemcit 30% less than the doses described herein. abine, 5FU)) and/or a platinum-based agent (e.g., carbopl I0082 In one aspect, the disclosure features, a method of atin, cisplatin, oxaliplatin). treating a proliferative disorder, e.g., a cancer, in a Subject. 0079. In one embodiment, the subject has ovarian cancer The method comprises: that is refractory, relapsed or resistant to a platinum-based I0083 providing an initial administration of a CDP-topoi agent, and the Subject is administered a CDP-topoisomerase Somerase inhibitor conjugate, particle or composition, e.g., a inhibitor conjugate, particle or composition, e.g., a CDP CDP-camptothecin or camptothecin derivative conjugate, topoisomerase inhibitor conjugate, particle or composition particle or composition, e.g., a CDP-camptothecin or camp described herein. In one embodiment, the CDP-topoi tothecin derivative conjugate, particle or composition Somerase inhibitor conjugate, particle or composition is described herein, e.g., CRLX101, to said Subject at a dosage administered in combination with doxorubicin (e.g., liposo of 3 mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m. mal doxorubicin). In one embodiment, the doxorubicin (e.g., 9 mg/m, 10 mg/m, or 11 mg/m, (wherein said dosage is the liposomal doxorubicin) is administered at a dose of about expressed in mg of drug, as opposed to mg of conjugate), 20 mg/m, about 30 mg/m or about 40 mg/m, every 24, 25, I0084 optionally, providing one or more subsequent 26, 27, 28, 29, 30 or 31 days, e.g., 28 days. In one embodi administrations of said CDP-topoisomerase inhibitor conju ment, the CDP-topoisomerase inhibitor conjugate, particle or gate, particle or composition, e.g., a CDP-camptothecin or US 2011/O160 159 A1 Jun. 30, 2011 camptothecin derivative conjugate, particle or composition, mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 e.g., a CDP-camptothecin or camptothecin derivative conju mg/m, 10 mg/m, or 11 mg/m, and gate, particle or composition described herein, e.g., 0093 one or more subsequent administrations of CRLX101, at a dosage of 3 mg/m, 4 mg/m, 5 mg/m, 6 CRLX101 to said subject, at a dosage of 3 mg/m, 4 mg/m. mg/m, 7 mg/m, 8 mg/m.9 mg/m, 10 mg/m, or 11 mg/m. 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m.9 mg/m, 10 mg/m. wherein each Subsequent administration is provided, inde or 11 mg/m, e.g., at the same dosage as the initial dosage, pendently, between 5, 6, 7, 8, 9 days after the previous, e.g., wherein each Subsequent administration is administered, the initial, administration, to thereby treat the proliferative independently, 5-9, e.g., 7 days after the previous, e.g., the disorder. initial, administration, and the cancer is, e.g., lung cancer, 0085. In an embodiment, the dosage of at least 2, 3, 4, 5, 6, e.g., non-Small cell lung cancer and/or Small cell lung cancer 7, 8, 9, 10, 12, 15, or 20 administrations is the same. (e.g., squamous cell non-small cell lung cancer or squamous 0.086. In an embodiment, the time between at least 2, 3, 4, cell Small cell lung cancer). In one embodiment, the lung 5, 6, 7, 8, 9, 10, 12, 15, or administrations is the same. cancer is refractory, relapsed or resistant to a platinum based 0087. In an embodiment, each subsequent administration agent (e.g., carboplatin, cisplatin oxaliplatin) and/or a taxane is administered 5-9, e.g., 7 days after the previous adminis (e.g., docetaxel, paclitaxel, larotaxel, cabazitaxel). tration. In an embodiment, 3 administrations are given 0094. In an embodiment, the method includes an initial between 5, 6, 7, 8 or 9 days from the previous administration administration of CRLX101 to said subject at a dosage of 3 and the fourth administration is given between 10, 11, 12, 13, mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 14, 15 or 16 days from the previous administration. This mg/m, 10 mg/m, or 11 mg/m, and dosing schedule can be repeated with 3 additional adminis 0.095 one or more subsequent administrations of trations given between 5, 6, 7, 8 or 9 days from the previous CRLX101 to said subject, at a dosage of 3 mg/m, 4 mg/m. administration and the Subsequent administration given 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m.9 mg/m, 10 mg/m. between 10, 11, 12, 13, 14, 15 or 16 days from the previous or 11 mg/m, e.g., at the same dosage as the initial dosage, administration. wherein each Subsequent administration is administered, 0088. In an embodiment, at least 2,3,4,5,6,7,8,9, 10, 12, independently, 5-9, e.g., 7 days after the previous, e.g., the 15, 20, 50 or 100 administrations are administered to said initial, administration, and the cancer is, e.g., ovarian cancer. Subject. In one embodiment, the ovarian cancer is refractory, relapsed 0089. In an embodiment, the drug is provided at 9-33 or resistant to a platinum-based agent (e.g., carboplatin, cis mg/m/month. platin, oxaliplatin). In one embodiment, the CRLX101 is 0090. In an embodiment, the conjugate includes a topoi administered by intraperitoneal administration. somerase I inhibitor and/or a topoisomerase II inhibitor. In an 0096. In an embodiment, the method includes an initial embodiment, the conjugate includes atopoisomerase I inhibi administration of CRLX101 to said subject at a dosage of 3 tor or combination of topoisomerase I inhibitors, e.g., camp mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 tothecin, irinotecan, SN-38, topotecan, lamellarin D and mg/m, 10 mg/m, or 11 mg/m, and derivatives thereof. In an embodiment, the conjugate includes 0097 one or more subsequent administrations of a topoisomerase II inhibitor or a combination of topoi CRLX101 to said subject, at a dosage of 3 mg/m, 4 mg/m. Somerase II inhibitors, e.g., eptoposide, tenoposide, doxoru 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 mg/m, 10 mg/m, bicin and derivatives thereof. In one embodiment, the conju or 11 mg/m, e.g., at the same dosage as the initial dosage, gate includes a combination of one or more topoisomerase I wherein each Subsequent administration is administered, inhibitors and one or more topoisomerase II inhibitors. In an independently, 5-9, e.g., 7 days after the previous, e.g., the embodiment, the CDP-topoisomerase inhibitor conjugate is a CDP-camptothecin or camptothecin derivate conjugate, e.g., initial, administration, and the cancer is, e.g., gastric cancer, a CDP-camptothecin or camptothecin derivative conjugate e.g., gastroesophageal, upper gastric or lower gastric cancer. described herein, e.g., CRLX101. 0098. In an embodiment, the method includes an initial 0091. In one embodiment, the CDP-topoisomerase inhibi administration of CRLX101 to said subject at a dosage of 3 tor conjugate, particle or composition, e.g., a CDP-camptoth mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 ecin or camptothecin derivative, a CDP-camptothecin or mg/m, 10 mg/m, or 11 mg/m, and camptothecin derivative conjugate, particle or composition 0099 one or more subsequent administrations of described herein, e.g., CRLX101, is administered by intrave CRLX101 to said subject, at a dosage of 3 mg/m, 4 mg/m. nous administration over a period equal to or less than about 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 mg/m, 10 mg/m, 30 minutes, 45 minutes, 60 minutes, 90 minutes, 120 minutes, or 11 mg/m, e.g., at the same dosage as the initial dosage, 150 minutes, or 180 minutes. In one embodiment, the CDP wherein each Subsequent administration is administered, topoisomerase inhibitor conjugate, particle or composition, independently, 5-9, e.g., 7 days after the previous, e.g., the e.g., a CDP-camptothecin or camptothecin derivative conju initial, administration, and the cancer is, e.g., pancreatic can gate, particle or composition, e.g., the CDP-camptothecin or C. camptothecin derivative conjugate, particle or composition 0100. In an embodiment, the method includes an initial described herein, e.g. CRLX101, is administered at a dosage administration of CRLX101 to said subject at a dosage of 3 of 3 mg/m, 4 mg/m.5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m. mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 9 mg/m, 10 mg/m, or 11 mg/m by intravenous administra mg/m, 10 mg/m, or 11 mg/m, and tion over a period equal to or less than about 30 minutes, 45 0101 one or more subsequent administrations of minutes, 60 minutes or 90 minutes, e.g., a period equal to or CRLX101 to said subject, at a dosage of 3 mg/m, 4 mg/m. less than 30 minutes, 45 minutes or 60 minutes. 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 mg/m, 10 mg/m, 0092. In an embodiment, the method includes an initial or 11 mg/m, e.g., at the same dosage as the initial dosage, administration of CRLX101 to said subject at a dosage of 3 wherein each Subsequent administration is administered, US 2011/O160 159 A1 Jun. 30, 2011 independently, 5-9, e.g., 7 days after the previous, e.g., the tothecin or camptothecin derivative conjugate, particle or initial, administration, and the cancer is, e.g., colorectal can composition described herein, e.g., CRLX101, prior to the C. initial administration. 0102. In an embodiment, the method includes an initial 0107. In an embodiment, the CDP-topoisomerase inhibi administration of CRLX101 to said subject at a dosage of 3 tor conjugate, particle or composition is administered as a mg/m, 4 mg/m, 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m, 9 first line treatment for the cancer. mg/m, 10 mg/m, or 11 mg/m, and 0108. In an embodiment, the CDP-topoisomerase inhibi tor conjugate, particle or composition is administered as a 0103 one or more subsequent administrations of second, third or fourth line treatment for the cancer. In an CRLX101 to said subject, at a dosage of 3 mg/m, 4 mg/m. embodiment, the cancer is sensitive to one or more chemo 5 mg/m, 6 mg/m, 7 mg/m, 8 mg/m.9 mg/m, 10 mg/m. therapeutic agents, e.g., a platinum-based agent, a taxane, an or 11 mg/m, e.g., at the same dosage as the initial dosage, alkylating agent, an antimetabolite and/or a Vinca alkaloid. In wherein each Subsequent administration is administered, an embodiment, the cancer is a refractory, relapsed or resis independently, 5-9, e.g., 7 days after the previous, e.g., the tant to initial, administration, and the cancer is, e.g., breast cancer, 0109 one or more chemotherapeutic agents, e.g., a plati e.g., estrogen receptor positive breast cancer, estrogen recep num-based agent, a taxane, an alkylating agent, an anthracy tor negative breast cancer, HER-2 positive breast cancer, cline (e.g., doxorubicin (e.g., liposomal doxorubicin)), and an HER-2 negative breast cancer, triple negative breast cancer or antimetabolite and/or a Vinca alkaloid. In one embodiment, inflammatory breast cancer. the cancer is, e.g., ovarian cancer, and the ovarian cancer is 0104. In an embodiment, the cancer is a cancer described refractory, relapsed or resistant to a platinum-based agent herein. For example, the cancer can be a cancer of the bladder (e.g., carboplatin, cisplatin, oxaliplatin), a taxane (e.g., pacli (including accelerated and metastatic bladder cancer), breast taxel, docetaxel, larotaxel, cabazitaxel) and/or an anthracy (e.g., estrogen receptor positive breast cancer, estrogen recep cline (e.g., doxorubicin (e.g., liposomal doxorubicin)). In one tor negative breast cancer, HER-2 positive breast cancer, embodiment, the cancer is, e.g., colorectal cancer, and the HER-2 negative breast cancer, triple negative breast cancer, cancer is refractory, relapsed or resistant to an antimetabolite (e.g., an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) inflammatory breast cancer), colon (including colorectal can and a pyrimidine analogue (e.g., capecitabine, cytrarabine, cer), kidney, liver, lung (including Small cell lung cancer and gemcitabine, 5FU)) and/or a platinum-based agent (e.g., car non-Small cell lung cancer (including adenocarcinoma, squa boplatin, cisplatin, oxaliplatin). In one embodiment, the can mous cell carcinoma, bronchoalveolar carcinoma and large cer is, e.g., lung cancer, and the cancer is refractory, relapsed cell carcinoma)), genitourinary tract, e.g., ovary (including or resistant to a taxane (e.g., paclitaxel, docetaxel, larotaxel, fallopian, endometrial and peritoneal cancers), cervix, pros cabazitaxel), a platinum-based agent (e.g., carboplatin, cispl tate and testes, lymphatic system, rectum, larynx, pancreas atin, oxaliplatin), a Vinca alkaloid (e.g., vinblastine, Vincris (including exocrine pancreatic carcinoma), stomach (e.g., tine, Vindesine, Vinorelbine), a vascular endothelial growth gastroesophageal, upper gastric or lower gastric cancer), gas factor (VEGF) pathway inhibitor, an epidermal growth factor trointestinal cancer (e.g., anal cancer), gallbladder, thyroid, (EGF) pathway inhibitor) and/or an antimetabolite (e.g., an lymphoma (e.g., Burkitt's, Hodgkin’s or non-Hodgkin's lym antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a phoma), leukemia (e.g., acute myeloid leukemia), Ewing's pyrimidine analogue (e.g., capecitabine, cytrarabine, gemcit sarcoma, nasoesophageal cancer, nasopharyngeal cancer, abine, 5FU)). In one embodiment, the cancer is, e.g., breast neural and glial cell cancers (e.g., glioblastoma multiforme), cancer, and the cancer is refractory, relapsed or resistant to a and head and neck. Preferred cancers include breast cancer taxane (e.g., paclitaxel, docetaxel, larotaxel, cabazitaxel), a (e.g., metastatic or locally advanced breast cancer), prostate vascular endothelial growth factor (VEGF) pathway inhibi cancer (e.g., hormone refractory prostate cancer), renal cell tor, an anthracycline (e.g., daunorubicin, doxorubicin (e.g., carcinoma, lung cancer (e.g., Small cell lung cancer and non liposomal doxorubicin), epirubicin, valrubicin, idarubicin), a Small cell lung cancer (including adenocarcinoma, squamous platinum-based agent (e.g., carboplatin, cisplatin, oxalipl cell carcinoma, bronchoalveolar carcinoma and large cell atin) and/or an antimetabolite (e.g., an antifolate (e.g., pem carcinoma)), pancreatic cancer, gastric cancer (e.g., gastroe etrexed, floxuridine, raltitrexed) and a pyrimidine analogue sophageal, upper gastric or lower gastric cancer), colorectal (e.g., capecitabine, cytrarabine, gemcitabine, 5FU)). In one cancer, Squamous cell cancer of the head and neck, ovarian embodiment, the cancer is, e.g., gastric cancer, and the cancer cancer (e.g., advanced ovarian cancer, platinum-based agent is refractory, relapsed or resistant to an antimetabolite (e.g., resistant or relapsed ovarian cancer), lymphoma (e.g., Bur an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a kitt's, Hodgkin's or non-Hodgkin’s lymphoma), leukemia pyrimidine analogue (e.g., capecitabine, cytrarabine, gemcit (e.g., acute myeloid leukemia) and gastrointestinal cancer. abine, 5FU)) and/or a platinum-based agent (e.g., carbopl 0105. In an embodiment, the cancer is ovarian, colorectal, atin, cisplatin, oxaliplatin). breast, lung, lymphoma orgastric cancer. In an embodiment, 0110. In one embodiment, the subject has ovarian cancer the cancer is a cancer other than pancreatic cancer, renal cell that is refractory, relapsed or resistant to a platinum-based carcinoma and/or lung cancer (e.g., non-Small cell lung can agent, and the Subject is administered a CDP-topoisomerase cer). In an embodiment, the cancer is a cancer other than inhibitor conjugate, particle or composition, e.g., a CDP pancreatic cancer, renal cell carcinoma, lung cancer (e.g., topoisomerase inhibitor conjugate, particle or composition non-Small cell lung cancer) and/or ovarian cancer. described herein. In one embodiment, the CDP-topoi 0106. In one embodiment, the subject has not been admin Somerase inhibitor conjugate, particle or composition is istered a CDP-topoisomerase inhibitor conjugate, particle or administered in combination with doxorubicin (e.g., liposo composition, e.g., a CDP-camptothecin or camptothecin mal doxorubicin). In one embodiment, the doxorubicin (e.g., derivative conjugate, particle or composition, a CDP-camp the liposomal doxorubicin) is administered at a dose of about US 2011/O160 159 A1 Jun. 30, 2011

20 mg/m, about 30 mg/m or about 40 mg/m, every 24, 25, 0.115. In one embodiment, the CDP-topoisomerase inhibi 26, 27, 28, 29, 30 or 31 days, e.g., 28 days. In one embodi tor conjugate, particle or composition, e.g., a CDP-camptoth ment, when the CDP-topoisomerase inhibitor conjugate, par ecin or camptothecin derivative conjugate, particle or com ticle or composition is administered in combination with position, e.g., a CDP-camptothecin or camptothecin doxorubicin (e.g., liposomal doxorubicin), the dose at which derivative conjugate, particle or composition described the CDP-topoisomerase inhibitor conjugate, particle or com herein, e.g., CRLX101, is administered prior to Surgery, after position is administered is 1%, 3%. 5%, 10%, 15%, 20%, Surgery or before and after Surgery to remove the cancer, e.g., 25%, 30% less than a dose described herein. to remove a primary tumor and/or a metastases. 0116. In one embodiment, the method comprises admin 0111. In an embodiment, the cancer has been sensitized to istering a CDP-topoisomerase inhibitor conjugate, particle or atopoisomerase inhibitor, e.g., the Subject has received radia composition, e.g., a CDP-camptothecin or camptothecin tion and/or the subject has received a phosphatase inhibitor derivative conjugate, particle or composition, e.g., a CDP (e.g., okadiac acid) prior to the administration of the CDP camptothecin or camptothecin derivative conjugate, particle topoisomerase inhibitor conjugate, particle or composition. or composition described herein, e.g., CRLX101, in combi In an embodiment, the cancer is sensitized to topoisomerase nation with a platinum-based agent (e.g., cisplatin, carbopl inhibitors, e.g., the Subject receives radiation in combination atin, oxaliplatin). In one embodiment, the conjugate, particle with the administration of the CDP-topoisomerase inhibitor or composition is further administered in combination with conjugate, particle or composition and/or the Subject is an anti-metabolite, e.g., an antifolate (e.g., pemetrexed, administered a phosphatase inhibitor (e.g., okadiac acid) in floXuridine, raltitrexed) or pyrimidine analogue (e.g., capecit combination with the administration of the CDP-topoi abine, cytrarabine, gemcitabine, 5FU)). In one embodiment, Somerase inhibitor conjugate, particle or composition. In one the conjugate, particle or composition is further administered embodiment, the cancer is sensitized or has been sensitized to in combination with an anti-metabolite, e.g., an antifolate topoisomerase inhibitors and the cancer is a glial cell cancer (e.g., pemetrexed, floXuridine, raltitrexed) or pyrimidine ana (e.g., glioblastoma multiforme) or head and neck cancer. logue (e.g., capecitabine, cytrarabine, gemcitabine, 5FU)) 0112 In one embodiment, the conjugate, particle or com and folinic acid (leucovorin). position is administered in combination with one or more 0117. In one embodiment, the conjugate, particle or com additional chemotherapeutic agent, e.g., a chemotherapeutic position is administered in combination with an angiogenesis agent (such as an angiogenesis inhibitor), or combination of inhibitor (e.g., an angiogenesis inhibitor described herein chemotherapeutic agents described herein. In one embodi such as an inhibitor of the VEGF pathway). In one embodi ment, the composition is administered in combination with ment, the method comprises administering a CDP-topoi one or more of a platinum-based agent (e.g., carboplatin, Somerase inhibitor conjugate, particle or composition, e.g., a cisplatin, oxaliplatin), a taxane (e.g., paclitaxel, docetaxel, CDP-camptothecin or camptothecin derivative conjugate, larotaxel, cabazitaxel), a Vinca alkaloid (e.g., vinblastine, Vin particle or composition, e.g., a CDP-camptothecin or camp cristine, Vindesine, Vinorelbine), an antimetabolite (e.g., an tothecin derivative conjugate, particle or composition antifolate (e.g., pemetrexed, floXuridine, raltitrexed) and a described herein, e.g., CRLX101, in combination with a tax pyrimidine analogue (e.g., 5FU, capecitabine, cytrarabine, ane (e.g., paclitaxel, docetaxel, larotaxel, cabazitaxel). In one gemcitabine)), an alkylating agent (e.g., cyclophosphamide, embodiment, the conjugate, particle or composition is further decarbazine, melphalan, ifosfamide, temozolomide), a vas administered in combination with a platinum-based agent cular endothelial growth factor (VEGF) pathway inhibitor, a (e.g., cisplatin, carboplatin, oxaliplatin). In one embodiment, poly ADP-ribose polymerase (PARP) inhibitor and an mTOR the conjugate, particle or composition is further administered inhibitor. In one embodiment, when the CDP-topoisomerase in combination with an anti-metabolite, e.g., an antifolate inhibitor conjugate, particle or composition is administered (e.g., pemetrexed, floXuridine, raltitrexed) or pyrimidine ana in combination with an additional chemotherapeutic agent, logue (e.g., capecitabine, cytrarabine, gemcitabine, 5FU)). In the dose at which the CDP-topoisomerase inhibitor conju one embodiment, the conjugate, particle or composition is gate, particle or composition is administered is 1%, 3%. 5%, further administered in combination with an anti-metabolite, 10%, 15%, 20%, 25%, 30% less thana dose described herein. e.g., an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) 0113. In an embodiment, the method further comprises or pyrimidine analogue (e.g., capecitabine, cytrarabine, gem administering to said subject a treatment that reduces one or citabine, 5FU)) and folinic acid (leucovorin). more side effect associated with administration of a CDP 0118. In one embodiment, the method comprises admin topoisomerase inhibitor conjugate, particle or composition, istering a CDP-topoisomerase inhibitor conjugate, particle or e.g., a treatment described herein. composition, e.g., a CDP-camptothecin or camptothecin 0114. In one aspect, the disclosure features, a method of derivative conjugate, particle or composition, e.g., a CDP treating ovarian cancer (e.g., epithelial carcinoma, fallopian camptothecin or camptothecin derivative conjugate, particle tube cancer, germ cell cancer (e.g., a teratoma), sex cord or composition described herein, e.g., CRLX101, in combi stromal tumor (e.g., estrogen-producing granulose cell nation with an anthracycline (e.g., doxorubicin (e.g., liposo tumor, virilizing Sertoli-Leydig tumor, arrhenoblastoma)), mal doxorubicin), daunorubicin, epirubicin, idarubicin, e.g., locally advanced or metastatic ovarian cancer, in a Sub , valrubicin). In one embodiment, the cancer is ject, e.g., a human Subject. The method comprises adminis refractory, relapsed or resistant to a taxane and/or a platinum tering a CDP-topoisomerase inhibitor conjugate, particle or based agent. composition, e.g., a CDP-camptothecin or camptothecin 0119. In one embodiment, the conjugate, particle or com derivative conjugate, particle or composition, e.g., a CDP position is administered in combination with one or more of camptothecin or camptothecin derivative conjugate, particle an anti-metabolite, e.g., an antifolate (e.g., pemetrexed, or composition described herein, e.g., CRLX101, in combi floXuridine, raltitrexed) or pyrimidine analogue (e.g., capecit nation with a second chemotherapeutic agent. abine, cytrarabine, gemcitabine, 5FU); an alkylating agent US 2011/O160 159 A1 Jun. 30, 2011 11

(e.g., cyclophosphamide, decarbazine, melphalan, ifosfa 0.125. In one embodiment, the conjugate, particle or com mide, temozolomide); a platinum-based agent (carboplatin, position is administered in combination with an angiogenesis cisplatin, oxaliplatin); a Vinca alkaloid (e.g., vinblastine, Vin inhibitor (e.g., an angiogenesis inhibitor described herein cristine, Vindesine, Vinorelbine). In one embodiment, the con such as an inhibitor of the VEGF pathway). In one embodi jugate, particle or composition is administered in combina ment, the method includes administering a CDP-topoi tion with one or more of capecitabine, cyclophosphamide, Somerase inhibitor conjugate, particle or composition, e.g., a gemcitabine, ifosfamide, melphalan, oxaliplatin, vinorelbine, CDP-camptothecin or camptothecin derivative conjugate, Vincristine and pemetrexed. In one embodiment, the cancer is particle or composition, e.g., a CDP-camptothecin or camp refractory, relapsed or resistant to a taxane and/or a platinum tothecin derivative conjugate, particle or composition described herein, e.g., CRLX101, in combination with a plati based agent. num-based agent (e.g., cisplatin, carboplatin, oxaliplatin). 0120 In one embodiment, the conjugate, particle or com I0126. In one embodiment, the method comprises admin position is administered at a dose and/or dosing schedule istering a CDP-topoisomerase inhibitor conjugate, particle or described herein. In one embodiment, when the CDP-topoi composition, e.g., a CDP-camptothecin or camptothecin Somerase inhibitor conjugate, particle or composition is derivative conjugate, particle or composition, e.g., a CDP administered in combination with an additional chemothera camptothecin or camptothecin derivative conjugate, particle peutic agent, the dose at which the CDP-topoisomerase or composition described herein, e.g., CRLX101, in combi inhibitor conjugate, particle or composition is administered is nation with a vascular endothelial growth factor (VEGF) 1%, 3%, 5%, 10%, 15%, 20%, 25%, 30% less than a dose pathway inhibitor, e.g., a VEGF inhibitor or VEGF receptor described herein. inhibitor. In one embodiment, the VEGF inhibitor is bevaci 0121. In one embodiment, the conjugate, particle or com Zumab or AV-951. In one embodiment, the VEGF receptor position is administered in combination with a treatment that inhibitor is selected from CP-547.632 and AZD2171. In one reduces one or more side effect associated with the adminis embodiment, the conjugate, particle or composition is admin tration of a CDP-topoisomerase inhibitor conjugate, particle istered in combination with a VEGF pathway inhibitor, e.g., or composition, e.g., a treatment described herein. bevacizumab, and an antimetabolite, e.g., an antifolate (e.g., 0122. In another aspect the disclosure features a method of pemetrexed, floruridine, raltitrexed) or pyrimidine analogue treating colorectal cancer (e.g., colon, Small intestine, rectum (e.g., capecitabine, 5FU, cytrarabine, gemcitabine). In one and/or appendix cancer), e.g., locally advanced or metastatic embodiment, the conjugate, particle or composition is admin colorectal cancer, in a subject, e.g., a human subject. The istered with a VEGF pathway inhibitor, e.g., bevacizumab, an method comprises administering a CDP-topoisomerase antimetabolite, e.g., a pyrimidine analogue (e.g., 5FU), and inhibitor conjugate, particle or composition, e.g., a CDP folinic acid (leucovorin). In another embodiment, the conju camptothecin or camptothecin derivative conjugate, particle gate, particle or composition is administered with a VEGF or composition, e.g., a CDP-camptothecin or camptothecin pathway inhibitor, e.g., bevacizumab, an antimetabolite, e.g., derivative conjugate, particle or composition described a pyrimidine analogue (e.g., 5FU), folinic acid (leucovorin), herein, e.g., CRLX101, in combination with a second chemo and a platinum-based agent (e.g., cisplatin, carboplatin, therapeutic agent. oxaliplatin). In one embodiment, the cancer is refractory, 0123. In one embodiment, the CDP-topoisomerase inhibi relapsed or resistant to an antimetabolite and/or a platinum tor conjugate, particle or composition, e.g., a CDP-camptoth based agent. ecin or camptothecin derivative conjugate, particle or com I0127. In another embodiment, a CDP-topoisomerase position, e.g., a CDP-camptothecin or camptothecin inhibitor conjugate, particle or composition, e.g., a CDP derivative conjugate, particle or composition described camptothecin or camptothecin derivative conjugate, particle herein, e.g., CRLX101, is administered prior to Surgery, after or composition, e.g., a CDP-camptothecin or camptothecin Surgery or before and after Surgery to remove the cancer, e.g., derivative conjugate, particle or composition described to remove the primary tumor and/or a metastases. herein, e.g., CRLX101, is administered in combination with a 0.124. In one embodiment, the method comprises admin VEGF pathway inhibitor, e.g., bevacizumab, and an antime istering a CDP-topoisomerase inhibitor conjugate, particle or tabolite wherein the antimetabolite is a pyrimidine analogue, composition, e.g., a CDP-camptothecin or camptothecin e.g., capecitabine. In one embodiment, the conjugate, particle derivative conjugate, particle or composition, e.g., a CDP or composition is further administered in combination with a camptothecin or camptothecin derivative conjugate, particle platinum-based agent (e.g., cisplatin, carboplatin, oxalipl or composition described herein, e.g., CRLX101, in combi atin). For example, in one embodiment, the conjugate, par nation with an antimetabolite, e.g., an antifolate (e.g., pem ticle or composition is administered with the following com etrexed, floxuridine, raltitrexed). In one embodiment, the bination: a VEGF pathway inhibitor, e.g., a VEGF inhibitor conjugate, particle or composition is administered in combi (e.g., bevacizumab) or a VEGF receptor inhibitor, a pyrimi nation with an antimetabolite, e.g., pyrimidine analogue (e.g., dine analogue (e.g., capecitabine), and a platinum-based capecitabine, cytrarabine, gemcitabine, 5FU) and folinic acid agent (e.g., Oxaliplatin); or a VEGF pathway inhibitor (e.g., (leucovorin). In one embodiment, the conjugate, particle or bevacizumab) and a pyrimidine analogue (e.g., capecitabine). composition is further administered in combination with a In one embodiment, the cancer is refractory, relapsed or resis platinum-based agent (e.g., cisplatin, carboplatin, oxalipl tant to an antimetabolite and/or a platinum-based agent. atin). For example, in one embodiment, the conjugate, par I0128. In one embodiment, a CDP-topoisomerase inhibitor ticle or composition is administered in combination with an conjugate, particle or composition, e.g., a CDP-camptothecin antimetabolite, e.g., 5FU, folinic acid (leucovorin), and a or camptothecin derivative conjugate, particle or composi platinum-based agent, e.g., Oxaliplatin. In another embodi tion, e.g., a CDP-camptothecin or camptothecin derivative ment, the antimetabolite is a pyrimidine analogue, e.g., conjugate, particle or composition described herein, e.g., capecitabine. CRLX101, is administered in combination with an epidermal US 2011/O160 159 A1 Jun. 30, 2011

growth factor (EGF) pathway inhibitor, e.g., an EGF inhibitor I0135) In one embodiment, the subject has increased or EGF receptor inhibitor. The EGF receptor inhibitor can be, KRAS and/or ST expression levels, e.g., as compared to a e.g., cetuximab, erlotinib, gefitinib, panitumumab. In one reference standard, and/or has a mutation in a KRAS and/or embodiment, the conjugate, particle or composition is admin ST gene. In one embodiment, the Subject has a mutation at istered in combination with an EGF pathway inhibitor, e.g., one or more of codon 12 of the KRAS gene (e.g., a G to T cetuximab or panitumumab, and a VEGF pathway inhibitor, transversion), codon 13 of the KRAS gene, codon 61 of the e.g., bevacizumab. In one embodiment, the cancer is refrac KRAS gene. In one embodiment, the Subject has non Small tory, relapsed or resistant to an antimetabolite and/or a plati cell lung cancer associated with mucinous broncholoalveolar num-based agent. cells or goblet cells. 0.136. In one embodiment, the method includes selecting a 0129. In one embodiment, the conjugate, particle or com Subject who has lung cancer and who has a mutation in an position is administered at a dose and/or dosing schedule EGFR gene; and described herein. In one embodiment, when the CDP-topoi 0.137 administering a CDP-topoisomerase inhibitor con Somerase inhibitor conjugate, particle or composition is jugate, particle or composition, e.g., a CDP-camptothecin or administered in combination with an additional chemothera camptothecin derivative conjugate, particle or composition, peutic agent, the dose at which the CDP-topoisomerase e.g., a CDP-camptothecin or camptothecin derivative conju inhibitor conjugate, particle or composition is administered is gate, particle or composition described herein, e.g., 1%, 3%, 5%, 10%, 15%, 20%, 25%, 30% less than a dose CRLX101, to the subject in an amount effective to treat the described herein. cancer, to thereby treat the cancer. 0130. In one embodiment, the conjugate, particle or com 0.138. In one embodiment, the subject has lung cancer that position is administered in combination with a treatment that is resistant, relapsed or refractory to an EGF pathway inhibi reduces one or more side effect associated with the adminis tor, e.g., an EGF receptor inhibitor (e.g., erlotinib) tration of a CDP-topoisomerase inhibitor conjugate, particle 0.139. In one embodiment, the subject has one or more of or composition, e.g., a treatment described herein. the following mutations: codon 719 of the EGFR gene (e.g., 0131. In one aspect, the disclosure features a method of a missense mutation that results in a glycine to cysteine, treating lung cancer (e.g., Small cell lung cancer or non-small alanine or serine substitution at codon 719 of the EGFR cell lung cancer (e.g., adenocarcinoma, squamous cell carci gene), codon 746 of the EGFR gene (e.g., a deletion of one or noma, bronchoalveolar carcinoma and large cell carcinoma)), more nucleic acids of codon 746 of the EGFR gene), codon e.g., locally advanced or metastatic lung cancer, in a subject, 747 of the EGFR gene (e.g., a deletion of one or more nucleic e.g., a human Subject. The method comprises administering a acids of codon 747 of the EGFR gene), codon 748 of the CDP-topoisomerase inhibitor conjugate, particle or compo EGFR gene (e.g., a deletion of one or more nucleic acids of sition, e.g., a CDP-topoisomerase I or II inhibitor conjugate, codon 748 of the EGFR gene), codon 749 of the EGFR gene particle or composition, e.g., a CDP-camptothecin or camp (e.g., a deletion of one or more nucleic acids of codon 749 of tothecin derivative conjugate, particle or composition, e.g., a the EGFR gene), codon 750 of the EGFR gene (e.g., a dele CDP-camptothecin or camptothecin derivative conjugate, tion of one or more nucleic acids of codon 750 of the EGFR particle or composition described herein, e.g., CRLX101. In gene), codon 858 of the EGFR gene (e.g., a missense muta one embodiment, the method comprises selecting a subject tion that results in a leucine to Substitution at codon that has squamous cell lung cancer for treatment with a CDP 858 of the EGFR gene), a deletion in exon 19 of the EGFR topoisomerase inhibitor conjugate, particle or composition, gene, and an insert mutation at exon 20 of the EGFR gene. e.g., a CDP-topoisomerase I or II inhibitor conjugate, particle 0140. In one embodiment, the subject has a mutation in the or composition, e.g., a CDP-camptothecin or camptothecin EGFR gene and has a mutation in the KRAS gene and/or derivative conjugate, particle or composition, e.g., a CDP overexpresses KRAS, e.g., as compared to a reference stan camptothecin or camptothecin derivative conjugate, particle dard. or composition described herein, e.g., CRLX101. 0.141. In one embodiment, the method includes selecting a 0.132. In one embodiment, the CDP-topoisomerase inhibi Subject who has lung cancer and who does not have a muta tor conjugate, particle or composition, e.g., a CDP-camptoth tion in an EGFR gene; and ecin or camptothecin derivative conjugate, particle or com 0.142 administering a CDP-topoisomerase inhibitor con position, e.g., a CDP-camptothecin or camptothecin jugate, particle or composition, e.g., a CDP-camptothecin or derivative conjugate, particle or composition described camptothecin derivative conjugate, particle or composition, herein, e.g., CRLX101, is administered prior to Surgery, after e.g., a CDP-camptothecin or camptothecin derivative conju Surgery or before and after Surgery to remove the cancer, e.g., gate, particle or composition described herein, e.g., to remove a primary tumor and/or a metastases. CRLX101, to the subject in an amount effective to treat the 0133. In one embodiment, the method includes selecting a cancer, to thereby treat the cancer. Subject who has lung cancer and who has increased KRAS 0143. In one embodiment, the method includes selecting a and/or ST expression levels, e.g., as compared to a reference Subject who has squamous cell lung cancer, and standard, and/or has a mutation in a KRAS and/or ST gene; 0144 administering a CDP-topoisomerase inhibitor con and jugate, particle or composition, e.g., a CDP-camptothecin or 0134) administering a CDP-topoisomerase inhibitor con camptothecin derivative conjugate, particle or composition, jugate, particle or composition, e.g., a CDP-camptothecin or e.g., a CDP-camptothecin or camptothecin derivative conju camptothecin derivative conjugate, particle or composition, gate, particle or composition described herein, e.g., e.g., a CDP-camptothecin or camptothecin derivative conju CRLX101, to the subject in an amount effective to treat the gate, particle or composition described herein, e.g., cancer, to thereby treat the cancer. CRLX101, to the subject in an amount effective to treat the 0145. In one embodiment, the subject does not have one or cancer, to thereby treat the cancer. more of the following mutations: codon 719 of the EGFR US 2011/O160 159 A1 Jun. 30, 2011

gene (e.g., a missense mutation that results in a glycine to 0154) In one embodiment, the method comprises admin cysteine, alanine or serine substitution at codon 719 of the istering a CDP-topoisomerase inhibitor conjugate, particle or EGFR gene), codon 746 of the EGFR gene (e.g., a deletion of composition, e.g., a CDP-camptothecin or camptothecin one or more nucleic acids of codon 746 of the EGFR gene), derivative conjugate, particle or composition, e.g., a CDP codon 747 of the EGFR gene (e.g., a deletion of one or more camptothecin or camptothecin derivative conjugate, particle nucleic acids of codon 747 of the EGFR gene), codon 748 of or composition described herein, e.g., CRLX101, in combi the EGFR gene (e.g., a deletion of one or more nucleic acids nation with an epidermal growth factor (EGF) pathway of codon 748 of the EGFR gene), codon 749 of the EGFR inhibitor, e.g., an EGF inhibitor or EGF receptor inhibitor. gene (e.g., a deletion of one or more nucleic acids of codon The EGF receptor inhibitor can be, e.g., cetuximab, erlotinib, 749 of the EGFR gene), codon 750 of the EGFR gene (e.g., a gefitinib, panitumumab. In an embodiment, the Subject has a deletion of one or more nucleic acids of codon 750 of the mutation at codon 858 of the gene encoding the EGF receptor, EGFR gene), codon 858 of the EGFR gene (e.g., a missense e.g., that results in a Substitution of a leucine to an arginine in mutation that results in a leucine to arginine Substitution at the EGF receptor. In one embodiment, the conjugate, particle codon 858 of the EGFR gene), a deletion in exon 19 of the or composition is administered in combination with an EGF EGFR gene, and an insert mutation at exon 20 of the EGFR pathway inhibitor, e.g., cetuximab, erlotinib, gefitinib, pani gene. tumumab, and radiation. In one embodiment, the conjugate, 0146 In one embodiment, the subject has a mutation in the particle or composition is administered in combination with KRAS gene and/or overexpresses KRAS, e.g., as compared an EGF pathway inhibitor, e.g., cetuximab, erlotinib, gefi to a reference standard, and does not have a mutation in the tinib, panitumumab, and one or more additional chemothera EGFR gene. peutic agents. For example, the chemotherapeutic agent can 0147 In one embodiment, the subject is refractory, be a platinum-based agent (e.g., cisplatin, carboplatin, oxali relapsed or resistant to one or more chemotherapeutic agents, platin), a taxane (e.g., paclitaxel, docetaxel, larotaxel, caba e.g., a platinum-based agent (e.g., carboplatin, cisplatin, Zitaxel), a Vinca alkaloid (e.g., vinblastine, Vincristine, Vin oxaliplatin) and/or an EGF pathway inhibitor, e.g., an EGF desine, Vinorelbine), an anti-metabolite, e.g., an antifolate inhibitor or and EGFR inhibitor, e.g., erlotinib. (e.g., pemetrexed, floXuridine, raltitrexed) or pyrimidine ana 0148. In one embodiment, the conjugate, particle or com logue (e.g., capecitabine, cytrarabine, gemcitabine, 5FU), position is administered at a dose and/or dosing schedule and combinations thereof. described herein. 0.155. In one embodiment, a CDP-topoisomerase inhibitor 0149. In one embodiment, the conjugate, particle or com conjugate, particle or composition, e.g., a CDP-camptothecin position is administered in combination with a treatment that or camptothecin derivative conjugate, particle or composi reduces one or more side effect associated with the adminis tion, e.g., a CDP-camptothecin or camptothecin derivative tration of a CDP-topoisomerase inhibitor conjugate, particle conjugate, particle or composition described herein, e.g., or composition, e.g., a treatment described herein. CRLX101, is administered in combination with a vascular 0150. In one aspect, the disclosure features a method of endothelial growth factor (VEGF) pathway inhibitor, e.g., a treating lung cancer (e.g., Small cell lung cancer and non VEGF inhibitor or VEGF receptor inhibitor. In one embodi Small cell lung cancer (e.g., adenocarcinoma, squamous cell ment, the VEGF inhibitor is bevacizumab or AV-951. In one carcinoma, bronchoalveolar carcinoma and large cell carci embodiment, the VEGF receptor inhibitor is selected from noma)), e.g., locally advanced or metastatic lung cancer, in a CP-547632 and AZD2171. In one embodiment, the conju Subject, e.g., a human Subject. The method comprises admin gate, particle or composition is administered in combination istering a CDP-topoisomerase inhibitor conjugate, particle or with a VEGF pathway inhibitor, e.g., bevacizumab, and radia composition, e.g., a CDP-topoisomerase I or II inhibitor con tion. In one embodiment, the conjugate, particle or composi jugate, particle or composition, e.g., a CDP-camptothecin or tion is administered in combination with a VEGF pathway camptothecin derivative conjugate, particle or composition, inhibitor, e.g., bevacizumab, and one or more additional che e.g., a CDP-camptothecin or camptothecin derivative conju motherapeutic agents. For example, the chemotherapeutic gate, particle or composition described herein, e.g., agent can be a platinum-based agent (e.g., cisplatin, carbopl CRLX101, in combination with a second chemotherapeutic atin, oxaliplatin), a taxane (e.g., paclitaxel, docetaxel, laro agent. taxel, cabazitaxel), a Vinca alkaloid (e.g., vinblastine, Vinc 0151. In one embodiment, the method includes selecting a ristine, Vindesine, Vinorelbine), an anti-metabolite, e.g., an Subject who has squamous cell lung cancer, and antifolate (e.g., pemetrexed, floXuridine, raltitrexed) or pyri 0152 administering a CDP-topoisomerase inhibitor con midine analogue (e.g., capecitabine, cytrarabine, gemcitab jugate, particle or composition, e.g., a CDP-camptothecin or ine, 5FU), and combinations thereof. In one embodiment, the camptothecin derivative conjugate, particle or composition, cancer is refractory, relapsed or resistant to one or more e.g., a CDP-camptothecin or camptothecin derivative conju chemotherapeutic agents, e.g., an EGF pathway inhibitor, gate, particle or composition described herein, e.g., e.g., erlotinib. CRLX101, to the subject in an amount effective to treat the 0156. In one embodiment, the conjugate, particle or com cancer, to thereby treat the cancer. position is administered in combination with an angiogenesis 0153. In one embodiment, the CDP-topoisomerase inhibi inhibitor (e.g., an angiogenesis inhibitor described herein tor conjugate, particle or composition, e.g., a CDP-camptoth such as an inhibitor of the VEGF pathway). In one embodi ecin or camptothecin derivative conjugate, particle or com ment, a CDP-topoisomerase inhibitor conjugate, particle or position, e.g., a CDP-camptothecin or camptothecin composition, e.g., a CDP-camptothecin or camptothecin derivative conjugate, particle or composition described derivative conjugate, particle or composition, e.g., a CDP herein, e.g., CRLX101, is administered prior to Surgery, after camptothecin or camptothecin derivative conjugate, particle Surgery or before and after Surgery to remove the cancer, e.g., or composition described herein, e.g., CRLX101, is admin to remove a primary tumor and/or a metastases. istered in combination with a platinum-based agent (e.g., US 2011/O160 159 A1 Jun. 30, 2011 cisplatin, carboplatin, oxaliplatin). In one embodiment, the e.g., a CDP-camptothecin or camptothecin derivative conju conjugate, particle or composition is further administered in gate, particle or composition, e.g., a CDP-camptothecin or combination with a taxane (e.g., paclitaxel, docetaxel, laro camptothecin derivative conjugate, particle or composition taxel, cabazitaxel), a Vinca alkaloid (e.g., vinblastine, Vinc described herein, e.g., CRLX101, in combination with a sec ristine, Vindesine, Vinorelbine) and/or an anti-metabolite, ond chemotherapeutic agent. e.g., an antifolate (e.g., pemetrexed, floXuridine, raltitrexed) 0163. In one embodiment, the CDP-topoisomerase inhibi or pyrimidine analogue (e.g., capecitabine, cytrarabine, gem tor conjugate, particle or composition, e.g., a CDP-camptoth citabine, 5FU). In one embodiment, the method further ecin or camptothecin derivative conjugate, particle or com includes administering radiation to the Subject. In one position, e.g., a CDP-camptothecin or camptothecin embodiment, the cancer is refractory, relapsed or resistant to derivative conjugate, particle or composition described one or more chemotherapeutic agents, e.g., an EGF pathway herein, e.g., CRLX101, is administered prior to Surgery, after inhibitor (e.g., erlonitib), a VEGF pathway inhibitor and/or a Surgery or before and after Surgery to remove the cancer, e.g., taxane. to remove a primary tumor and/or a metastases. 0157. In one embodiment, a CDP-topoisomerase inhibitor 0164. In one embodiment, the CDP-topoisomerase inhibi conjugate, particle or composition, e.g., a CDP-camptothecin tor conjugate, particle or composition, e.g., a CDP-camptoth or camptothecin derivative conjugate, particle or composi ecin or camptothecin derivative conjugate, particle or com tion, e.g., a CDP-camptothecin or camptothecin derivative position, e.g., a CDP-camptothecin or camptothecin conjugate, particle or composition described herein, e.g., derivative conjugate, particle or composition described CRLX101, is administered in combination with a taxane herein, e.g., CRLX101, is administered in combination with a (e.g., paclitaxel, docetaxel, larotaxel, cabazitaxel). In one HER-2 pathway inhibitor, e.g., a HER-2 inhibitor or a HER-2 embodiment, the method further includes administering receptor inhibitor. For example, the CDP-topoisomerase radiation to the Subject. In one embodiment, the cancer is inhibitor conjugate, particle or composition is administered refractory, relapsed or resistant to one or more chemothera with trastuzumab. peutic agents, e.g., an EGF pathway inhibitor (e.g., erlotinib), 0.165. In some embodiments, the CDP-topoisomerase a VEGF pathway inhibitor and/or a platinum-based agent. inhibitor conjugate, particle or composition, e.g., a CDP 0158. In one embodiment, a CDP-topoisomerase inhibitor camptothecin or camptothecin derivative conjugate, particle conjugate, particle or composition, e.g., a CDP-camptothecin or composition, e.g., a CDP-camptothecin or camptothecin or camptothecin derivative conjugate, particle or composi derivative conjugate, particle or composition described tion, e.g., a CDP-camptothecin or camptothecin derivative herein, e.g., CRLX101, is administered in combination with a conjugate, particle or composition described herein, e.g., vascular endothelial growth factor (VEGF) pathway inhibi CRLX101, is administered in combination with an anti-me tor, e.g., a VEGF inhibitor (e.g., bevacizumab) or VEGF tabolite, e.g., an antifolate (e.g., pemetrexed, floXuridine, receptor inhibitor (e.g., CP-547632 and AZD2171). In one raltitrexed). In one embodiment, the method further includes embodiment, the CDP-topoisomerase inhibitor conjugate, administering radiation to the Subject. In one embodiment, particle or composition is administered in combination with the cancer is refractory, relapsed or resistant to bevacizumab. In some embodiments, the method further 0159 one or more chemotherapeutic agents, e.g., an EGF comprises administering a taxane (e.g., paclitaxel, docetaxel, pathway inhibitor (e.g., erlotinib), a VEGF pathway inhibitor, larotaxel, cabazitaxel). In one embodiment, the method fur a taxane and/or a platinum-based agent. ther comprises administering a poly ADP-ribose polymerase 0160. In one embodiment, the conjugate, particle or com (PARP) inhibitor (e.g., BSI 201, (AZD-2281), ABT position is administered at a dose and/or dosing schedule 888, AG014699, CEP 9722, MK 4827, KU-0059436 described herein. In one embodiment, when the CDP-topoi (AZD2281), LT-673, 3-aminobenzamide). Somerase inhibitor conjugate, particle or composition is 0166 In some embodiments, the CDP-topoisomerase administered in combination with an additional chemothera inhibitor conjugate, particle or composition, e.g., a CDP peutic agent, the dose at which the CDP-topoisomerase camptothecin or camptothecin derivative conjugate, particle inhibitor conjugate, particle or composition is administered is or composition, e.g., a CDP-camptothecin or camptothecin 1%, 3%, 5%, 10%, 15%, 20%, 25%, 30% less than a dose derivative conjugate, particle or composition described described herein. herein, e.g., CRLX101, is administered in combination with a 0161 In one embodiment, the conjugate, particle or com platinum-based agent (e.g., cisplatin, carboplatin, oxalipl position is administered in combination with a treatment that atin). In some embodiments, the method further comprises reduces one or more side effect associated with the adminis administering a taxane (e.g., paclitaxel, docetaxel, larotaxel, tration of a CDP-topoisomerase inhibitor conjugate, particle cabazitaxel). In some embodiments, the method further com or composition, e.g., a treatment described herein. prises administering an mTOR inhibitor. Non-limiting 0162. In one aspect, the disclosure features a method of examples of mTOR inhibitors include rapamycin, everoli treating breast cancer (e.g., estrogen receptor positive breast mus, AP23573, CCI-779 and SDZ-RAD. In one embodiment, cancer; estrogen receptor negative breast cancer; HER-2 the method further comprises administering a PARP inhibitor positive breast cancer; HER-2 negative breast cancer, proges (e.g., BSI 201, Olaparib (AZD-2281), ABT-888, AG014699, terone receptor positive breast cancer, progesterone receptor CEP 9722, MK 4827, KU-0059436 (AZD2281), LT-673, negative breast cancer; estrogen receptor negative, HER-2 3-aminobenzamide). In some embodiments, the method fur negative and progesterone receptor negative breast cancer ther comprises administering a VEGF pathway inhibitor, e.g., (i.e., triple negative breast cancer)), e.g., locally advanced or a VEGF inhibitor (e.g., bevacizumab) or VEGF receptor metastatic breast cancer, in a subject, e.g., a human Subject. inhibitor (e.g., CP-547632 and AZD2171). The method comprises administering a CDP-topoisomerase 0167. In one embodiment, the conjugate, particle or com inhibitor conjugate, particle or composition, e.g., a topoi position is administered in combination with an angiogenesis Somerase I or II inhibitor conjugate, particle or composition, inhibitor (e.g., an angiogenesis inhibitor described herein US 2011/O160 159 A1 Jun. 30, 2011

such as an inhibitor of the VEGF pathway). In some embodi camptothecin or camptothecin derivative conjugate, particle ments, the CDP-topoisomerase inhibitor conjugate, particle or composition, e.g., a CDP-camptothecin or camptothecin or composition, e.g., a CDP-camptothecin or camptothecin derivative conjugate, particle or composition described derivative conjugate, particle or composition, e.g., a CDP herein, e.g., CRLX101, is administered in combination with camptothecin or camptothecin derivative conjugate, particle an anthracycline (e.g., daunorubicin, doxorubicin (liposomal or composition described herein, e.g., CRLX101, is admin doxorubicin), epirubicin, valrubicin and idarubicin) and an istered in combination with a taxane (e.g., paclitaxel, doc anti-metabolite (e.g., floxuridine, pemetrexed, 5FU). In one etaxel, larotaxel, cabazitaxel). In some embodiments, the embodiment, the cancer is refractory, relapsed or resistant to method further comprises administering an mTOR inhibitor. one or more chemotherapeutic agents, e.g., a HER-2 pathway Non-limiting examples of mTOR inhibitors include rapamy inhibitor, a VEGF pathway inhibitor, and/or a platinum-based cin, , AP23573, CCI-779 and SDZ-RAD. In one agent. embodiment, the method further comprises administering a 0172. In some embodiments, the CDP-topoisomerase PARP inhibitor (e.g., BSI 201, Olaparib (AZD-2281), ABT inhibitor conjugate, particle or composition, e.g., a CDP 888, AG014699, CEP 9722, MK 4827, KU-0059436 camptothecin or camptothecin derivative conjugate, particle (AZD2281), LT-673, 3-aminobenzamide). or composition, e.g., a CDP-camptothecin or camptothecin 0.168. In some embodiments, the CDP-topoisomerase derivative conjugate, particle or composition described inhibitor conjugate, particle or composition, e.g., a CDP herein, e.g., CRLX101, is administered in combination with camptothecin or camptothecin derivative conjugate, particle an mTOR inhibitor. Non-limiting examples of mTOR inhibi or composition, e.g., a CDP-camptothecin or camptothecin tors include rapamycin, everolimus, AP23573, CCI-779 and derivative conjugate, particle or composition described SDZ-RAD. In some embodiments, the method further com herein, e.g., CRLX101, is administered in combination with prises administering a PARP inhibitor (e.g., BSI 201, Ola an (e.g., ixabelipone, epothilone B, epothilone D. parib (AZD-2281), ABT-888, AG014699, CEP 9722, MK BMS310705, dehydelone, ZK-EPO). In some embodiments, 4827, KU-0059436 (AZD2281), LT-673, 3-aminobenza the method further comprises administering an mTOR inhibi mide). tor. Non-limiting examples of mTOR inhibitors include rapa 0173. In some embodiments, the CDP-topoisomerase mycin, everolimus, AP23573, CCI-779 and SDZ-RAD. In inhibitor conjugate, particle or composition, e.g., a CDP one embodiment, the method further comprises administer camptothecin or camptothecin derivative conjugate, particle ing a PARP inhibitor (e.g., BSI 201, Olaparib (AZD-2281), or composition, e.g., a CDP-camptothecin or camptothecin ABT-888, AG014699, CEP 9722, MK 4827, KU-0059436 derivative conjugate, particle or composition described (AZD2281), LT-673, 3-aminobenzamide). In some embodi herein, e.g., CRLX101, is administered in combination with a ments, the method further comprises administering a VEGF PARP inhibitor (e.g., BSI 201, Olaparib (AZD-2281), ABT pathway inhibitor, e.g., a VEGF inhibitor (e.g., bevacizumab) 888, AG014699, CEP 9722, MK 4827, KU-0059436 or VEGF receptor inhibitor (e.g., CP-547632 and AZD217 1). (AZD2281), LT-673, 3-aminobenzamide). In some embodiments, the method further includes adminis 0.174. In some embodiments, the CDP-topoisomerase tering one or more of an anthracycline (e.g., daunorubicin, inhibitor conjugate, particle or composition, e.g., a CDP doxorubicin (liposomal doxorubicin), epirubicin, valrubicin camptothecin or camptothecin derivative conjugate, particle and idarubicin) and/or an anti-metabolite (e.g., floXuridine, or composition, e.g., a CDP-camptothecin or camptothecin pemetrexed, 5FU). derivative conjugate, particle or composition described 0169. In some embodiments, the CDP-topoisomerase herein, e.g., CRLX101, is administered in combination with a inhibitor conjugate, particle or composition, e.g., a CDP pyrimidine analogue, e.g., a pyrimidine analogue described camptothecin or camptothecin derivative conjugate, particle herein (e.g., capecitabine). In some embodiments, the method or composition, e.g., a CDP-camptothecin or camptothecin further comprises administering a taxane (e.g., docetaxel, derivative conjugate, particle or composition described paclitaxel, larotaxel, cabazitaxel). In some embodiments, the herein, e.g., CRLX101, is administered in combination with method further comprises administering an epothilone (e.g., an anthracycline (e.g., daunorubicin, doxorubicin (liposomal ixabelipone, epothilone B, epothilone D, BMS310705, doxorubicin), epirubicin, valrubicin and idarubicin). In one dehydelone, ZK-EPO). embodiment, the cancer is refractory, relapsed or resistant to 0.175. In one embodiment, the conjugate, particle or com one or more chemotherapeutic agents, e.g., a HER-2 pathway position is administered at a dose and/or dosing schedule inhibitor, a VEGF pathway inhibitor, a taxane, an antimetabo described herein. In one embodiment, when the CDP-topoi lite and/or a platinum-based agent. Somerase inhibitor conjugate, particle or composition is 0170 In some embodiments, the CDP-topoisomerase administered in combination with an additional chemothera inhibitor conjugate, particle or composition, e.g., a CDP peutic agent, the dose at which the CDP-topoisomerase camptothecin or camptothecin derivative conjugate, particle inhibitor conjugate, particle or composition is administered is or composition, e.g., a CDP-camptothecin or camptothecin 1%, 3%, 5%, 10%, 15%, 20%, 25%, 30% less than a dose derivative conjugate, particle or composition described described herein. herein, e.g., CRLX101, is administered in combination with 0176). In one embodiment, the conjugate, particle or com an anti-metabolite, e.g., an antifolate (e.g., floXuridine, pem position is administered in combination with a treatment that etrexed) or pyrimidine analogue (e.g., 5FU)). In one embodi reduces one or more side effect associated with the adminis ment, the cancer is refractory, relapsed or resistant to one or tration of a CDP-topoisomerase inhibitor conjugate, particle more chemotherapeutic agents, e.g., a HER-2 pathway or composition, e.g., a treatment described herein. inhibitor, a VEGF pathway inhibitor, a taxane, an anthracy 0177. In one aspect, the disclosure features a method of cline and/or a platinum-based agent. treating gastric cancer (e.g., gastric adenocarcinoma (e.g., 0171 In some embodiments, the CDP-topoisomerase intestinal or diffuse), gastric lymphoma (e.g., MALT lym inhibitor conjugate, particle or composition, e.g., a CDP phoma), carcinoid stromal tumor), e.g., locally advanced or US 2011/O160 159 A1 Jun. 30, 2011 metastatic gastric cancer, in a Subject, e.g., a human Subject. dine, pemetrexed, raltitrexed) or pyrimidine analogue (e.g., The method comprises administering a CDP-topoisomerase 5FU, capecitabine, cytrarabine, gemcitabine). inhibitor conjugate, particle or composition, e.g., a CDP 0182. In some embodiments, the CDP-topoisomerase topoisomerase I or II inhibitor conjugate, particle or compo inhibitor conjugate, particle or composition, e.g., a CDP sition, e.g., a CDP-camptothecin or camptothecin derivative camptothecin or camptothecin derivative conjugate, particle conjugate, particle or composition, e.g., a CDP-camptothecin or composition, e.g., a CDP-camptothecin or camptothecin or camptothecin derivative conjugate, particle or composition derivative conjugate, particle or composition described described herein, e.g., CRLX101, in combination with a sec herein, e.g., CRLX101, is administered in combination with ond chemotherapeutic agent. an anti-metabolite, e.g., an antifolate (e.g., floXuridine, pem etrexed, raltitrexed) or pyrimidine analogue (e.g., capecitab 0178. In one embodiment, the gastric cancer is gastroe ine, 5FU, cytrarabine, gemcitabine). In one embodiment, the sophageal junction adenocarcinoma. method further includes administering a taxane (e.g., pacli 0179. In one embodiment, the CDP-topoisomerase inhibi taxel, docetaxel, larotaxel, cabazitaxel). For example, in one tor conjugate, particle or composition, e.g., a CDP-camptoth embodiment, the CDP-topoisomerase inhibitor conjugate, ecin or camptothecin derivative conjugate, particle or com particle or composition, e.g., a CDP-camptothecin or camp position, e.g., a CDP-camptothecin or camptothecin tothecin derivative conjugate, particle or composition, e.g., a derivative conjugate, particle or composition described CDP-camptothecin or camptothecin derivative conjugate, herein, e.g., CRLX101, is administered prior to Surgery, after particle or composition described herein, e.g., CRLX101, is Surgery or before and after Surgery to remove the cancer, e.g., administered in combination with an anti-metabolite, e.g., an to remove a primary tumor and/or a metastases. antifolate (e.g., floXuridine, pemetrexed, raltitrexed) or pyri 0180. In one embodiment, the CDP-topoisomerase inhibi midine analogue (e.g., capecitabine, 5FU, cytrarabine, gem tor conjugate, particle or composition, e.g., a CDP-camptoth citabine) and a taxane (e.g., paclitaxel, docetaxel, larotaxel, ecin or camptothecin derivative conjugate, particle or com cabazitaxel). position, e.g., a CDP-camptothecin or camptothecin 0183 In one embodiment, the conjugate, particle or com derivative conjugate, particle or composition described position is administered in combination with an angiogenesis herein, e.g., CRLX101, is administered in combination with inhibitor (e.g., an angiogenesis inhibitor described herein one or more of an anthracycline (e.g., daunorubicin, doxoru such as an inhibitor of the VEGF pathway). bicin (e.g., liposomal doxorubicin), epirubicin, valrubicin, 0184. In one embodiment, the CDP-topoisomerase inhibi mitoxatrone, and idarubicin), a platinum-based agent (e.g., tor conjugate, particle or composition, e.g., a CDP-camptoth cisplatin, carboplatin, oxaliplatin) and an anti-metabolite, ecin or camptothecin derivative conjugate, particle or com e.g., an antifolate (e.g., floXuridine, pemetrexed, raltitrexed) position, e.g., a CDP-camptothecin or camptothecin or pyrimidine analogue (e.g., 5FU, capecitabine, cytrarabine, derivative conjugate, particle or composition described gemcitabine)). For example, in one embodiment, the CDP herein, e.g., CRLX101, is administered in combination with topoisomerase inhibitor conjugate, particle or composition, radiation. e.g., a CDP-camptothecin or camptothecin derivative conju 0185. In some embodiments, the CDP-topoisomerase gate, particle or composition, e.g., a CDP-camptothecin or inhibitor conjugate, particle or composition, e.g., a CDP camptothecin derivative conjugate, particle or composition camptothecin or camptothecin derivative conjugate, particle described herein, e.g., CRLX101, is administered in combi or composition, e.g., a CDP-camptothecin or camptothecin nation with an anthracycline (e.g., daunorubicin, doxorubicin derivative conjugate, particle or composition described (e.g., liposomal doxorubicin), epirubicin, valrubicin, mitox herein, e.g., CRLX101, is administered in combination with a atrone and idarubicin), a platinum-based agent (e.g., cispl vascular endothelial growth factor (VEGF) pathway inhibi atin, carboplatin, oxaliplatin) and an anti-metabolite, e.g., an tor, e.g., a VEGF inhibitor (e.g., bevacizumab) or VEGF antifolate (e.g., floXuridine, pemetrexed, raltitrexed) or pyri receptor inhibitor (e.g., CP-547632 and AZD2171). In one midine analogue (e.g., 5FU, capecitabine, cytrarabine, gem embodiment, the CDP-topoisomerase inhibitor conjugate, citabine). In one embodiment, the CDP-topoisomerase particle or composition, e.g., a CDP-camptothecin or camp inhibitor conjugate, particle or composition, e.g., a CDP tothecin derivative conjugate, particle or composition, e.g., a camptothecin or camptothecin derivative conjugate, particle CDP-camptothecin or camptothecin derivative conjugate, or composition, e.g., a CDP-camptothecin or camptothecin particle or composition described herein, e.g., CRLX101, is derivative conjugate, particle or composition described administered in combination with bevacizumab. In one herein, e.g., CRLX101, is administered in combination with embodiment, the cancer is refractory, relapsed or resistant to an anthracycline (e.g., daunorubicin, doxorubicin (e.g., lipo one or more chemotherapeutic agents, e.g., an antimetabolite, Somal doxorubicin), epirubicin, valrubicin, mitoxatrone and a platinum-based agent and/or an anthracycline. idarubicin). In one embodiment, the cancer is refractory, 0186. In some embodiments, the CDP-topoisomerase relapsed or resistant to one or more chemotherapeutic agents, inhibitor conjugate, particle or composition, e.g., a CDP e.g., a platinum-based agent (e.g., cisplatin, carboplatin, camptothecin or camptothecin derivative conjugate, particle oxaliplatin). or composition, e.g., a CDP-camptothecin or camptothecin 0181. In another embodiment, the CDP-topoisomerase derivative conjugate, particle or composition described inhibitor conjugate, particle or composition, e.g., a CDP herein, e.g., CRLX101, is administered in combination with camptothecin or camptothecin derivative conjugate, particle an mTOR inhibitor. Non-limiting examples of mTOR inhibi or composition, e.g., a CDP-camptothecin or camptothecin tors include rapamycin, everolimus, AP23573, CCI-779 and derivative conjugate, particle or composition described SDZ-RAD. In one embodiment, the cancer is refractory, herein, e.g., CRLX101, is administered in combination with a relapsed or resistant to one or more chemotherapeutic agents, platinum-based agent (e.g., cisplatin, carboplatin, oxalipl e.g., an antimetabolite, a platinum-based agent and/or an atin) and an anti-metabolite, e.g., an antifolate (e.g., floxuri anthracycline. US 2011/O160 159 A1 Jun. 30, 2011

0187. In some embodiments, the CDP-topoisomerase (Novavea), ARQ 197 (ArOule), ASA404 (Novartis/Anti inhibitor conjugate, particle or composition, e.g., a CDP soma), Atiprimod (Callisto Pharmaceuticals), ATN-161 (At camptothecin or camptothecin derivative conjugate, particle tenuon), AV-412 (Aveo Pharmaceuticals), AV-951 (Aveo or composition, e.g., a CDP-camptothecin or camptothecin Pharmaceuticals), Avastin (Bevacizumab) (Genentech), derivative conjugate, particle or composition described AZD2171 (Cediranib/Recentin) (AstraZeneca), BAY herein, e.g., CRLX101, is administered in combination with a 57-9352 (Telatinib) (Bayer), BEZ235 (Novartis), BIBF1120 poly ADP-ribose polymerase (PARP) inhibitor (e.g., BSI (Boehringer Ingelheim Pharmaceuticals), BIBW 2992 (Boe 201, Olaparib (AZD-2281), ABT-888, AG014699, CEP hiringer Ingelheim Pharmaceuticals), BMS-275291 (Bristol 9722, MK 4827, KU-0059436 (AZD2281), LT-673, 3-ami Myers Squibb), BMS-582664 (Brivanib) (Bristol-Myers nobenzamide). In one embodiment, the cancer is refractory, Squibb), BMS-690514 (Bristol-Myers Squibb), Calcitriol, relapsed or resistant to one or more chemotherapeutic agents, CCI-779 (Torisel) (Wyeth), CDP-791 (ImClone Systems), e.g., an antimetabolite, a platinum-based agent and/or an Ceflatonin (Homoharringtonine/HHT) (ChemGenex Thera anthracycline. peutics), Celebrex () (Pfizer), CEP-7055 (Cepha 0188 In one embodiment, the conjugate, particle or com lon/Sanofi), CHIR-265 (Chiron Corporation), NGR-TNF, position is administered at a dose and/or dosing schedule COL-3 (Metastat) (Collagenex Pharmaceuticals), Combret described herein. In one embodiment, when the CDP-topoi astatin (Oxigene), CP-751,871 (Figitumumab) (Pfizer), Somerase inhibitor conjugate, particle or composition is CP-547,632 (Pfizer), CS-7017 (Daiichi Sankyo Pharma), administered in combination with an additional chemothera CT-322 (Angiocept) (Adnexus), Curcumin, Dalteparin peutic agent, the dose at which the CDP-topoisomerase (Fragmin) (Pfizer), Disulfuram (Antabuse), E7820 (Eisai inhibitor conjugate, particle or composition is administered is Limited), E7080 (Eisai Limited), EMD 121974 (Cilengitide) 1%, 3%, 5%, 10%, 15%, 20%, 25%, 30% less than a dose (EMD Pharmaceuticals), ENMD-1198 (EntreMed), ENMD described herein. 2076 (EntreMed), Endostar (Simcere), Erbitux (ImClone/ 0189 In one embodiment, the conjugate, particle or com Bristol-Myers Squibb), EZN-2208 (Enzon Pharmaceuticals), position is administered in combination with a treatment that EZN-2968 (Enzon Pharmaceuticals), GC1008 (Genzyme), reduces one or more side effect associated with the adminis Genistein, GSK1363089 (Foretinib) (GlaxoSmithKline), tration of a CDP-topoisomerase inhibitor conjugate, particle GW786034 (Pazopanib) (GlaxoSmithKline), GT-111 (Vas or composition, e.g., a treatment described herein. cular Biogenics Ltd.), IMC-1 121 B (Ramucirumab) (Im 0190. In one aspect, the invention features, a method of Clone Systems), IMC-18F1 (ImClone Systems), IMC-3G3 treating pancreatic cancer in a subject, the method compris (ImClone LLC), INCB007839 (Incyte Corporation), INGN ing, administering a CDP-topoisomerase inhibitor conjugate, 241 (Introgen Therapeutics), Iressa (ZD1839/Gefitinib), particle or composition to the Subject in combination with a LBH589 (Faridak/Panobinostst) (Novartis), Lucentis pyrimidine analogue (e.g., capecitabine, 5FU, cytrarabine, (Ranibizumab) (Genentech/Novartis), LY317615 (Enzastau gemcitabine). In an embodiment, the pyrimidine analogue is rin) (Eli Lilly and Company), Macugen (Pegaptainib) (Pfizer), gemcitabine. MEDI522 (Abegrin) (MedImmune), MLN518 (Tandutinib) 0191 In one aspect, the invention features, a method of (Millennium), Neovastat (AE941/Benefin) (Aeterna Zenta treating a proliferative disorder, e.g., a cancer, in a subject, ris), Nexavar (Bayer/Onyx), NM-3 (Genzyme Corporation), e.g., a human Subject. The method comprises: Noscapine (Cougar Biotechnology), NPI-2358 (Nereus Phar 0.192 providing a subject who has a proliferative disorder, maceuticals), OSI-930 (OSI), Palomid 529 (Paloma Pharma e.g., cancer, and ceuticals, Inc.), Panzem Capsules (2ME2) (EntreMed), Pan 0193 administering a composition that comprises a CDP Zem NCD (2ME2) (EntreMed), PF-02341066 (Pfizer), topoisomerase inhibitor conjugate, particle or composition, PF-04554878 (Pfizer), PI-88 (Progen Industries/Medigen e.g., a CDP-topoisomerase I or II inhibitor conjugate, particle Biotechnology), PKC412 (Novartis), Polyphenon E (Green or composition, e.g., a CDP-camptothecin or camptothecin Tea Extract) (Polypheno E International, Inc), PPI-2458 derivative conjugate, particle or composition, e.g., a CDP (Praecis Pharmaceuticals), PTC299 (PTC Therapeutics), camptothecin derivative conjugate, particle or composition PTK787 () (Novartis), PXD101 () described herein, e.g., CRLX101, to said subject in combi (CuraGen Corporation), RAD001 (Everolimus) (Novartis), nation with an angiogenesis inhibitor. RAF265 (Novartis), Regorafenib (BAY73-4506) (Bayer), 0194 In one embodiment, the cancer is renal cancer. Revlimid (Celgene), Retaane (Alcon Research), SN38 (Lipo 0.195. In one embodiment, the conjugate, particle or com somal) (Neopharm), SNS-032 (BMS-387032) (Sunesis), position is administered in combination with an angiogenesis SOM230 (Pasireotide) (Novartis), Squalamine (Genaera), inhibitor described herein such as a VEGF pathway inhibitor Suramin, Sutent (Pfizer), Tarceva (Genentech), TB-403 (e.g., a VEGF pathway inhibitor described herein). Exem (Thrombogenics), Tempostatin (Collard Biopharmaceuti plary angiogenesis inhibitors include the following: A6 (Ang cals), Tetrathiomolybdate (Sigma-Aldrich), TG 100801 strom Pharmacueticals), ABT-510 (Abbott Laboratories), (TargeCien), (Celgene Corporation), Tinzaparin ABT-627 () (Abbott Laboratories/Xinlay), ABT Sodium, TKI258 (Novartis), TRC093 (Tracon Pharmaceuti 869 (Abbott Laboratories), Actimid (CC4047, Pomalido cals Inc.), VEGF Trap () (Regeneron Pharmaceu mide) (Celgene Corporation), AdOVPEDF.11D (GenVec), ticals), VEGF Trap-Eye (Regeneron Pharmaceuticals), Veg ADH-1 (Exherin) (Adherex Technologies), AEE788 (Novar lin (Vasciene Therapeutics), (Millennium), tis), AG-013736 (Axitinib) (Pfizer), AG3340 (Prinomastat) XL184 (Exelixis), XL647 (Exelixis), XL784 (Exelixis), (Agouron Pharmaceuticals), AGX1053 (AngioGenex), XL820 (Exelixis), XL999 (Exelixis), ZD6474 (AstraZen AGX51 (AngioGenex), ALN-VSP (ALN-VSPO2) (Alnylam eca), (Merck), and ZSTK474. Pharmaceuticals), AMG 386 (Amgen), AMG706 (Amgen), 0196. In one embodiment, the conjugate, particle or com Apatinib (YN968D1) (Jiangsu Hengrui Medicine), AP23573 position is administered in combination with a treatment that (/MK8669) (Ariad Pharmaceuticals), AQ4N reduces one or more side effect associated with the adminis US 2011/O160 159 A1 Jun. 30, 2011 tration of a CDP-topoisomerase inhibitor conjugate, particle 0209 administering a composition that comprises a camp or composition, e.g., a treatment described herein. tothecin or camptothecin derivative, e.g., a CDP-topoi 0197) In one aspect, the disclosure features, a method of Somerase inhibitor conjugate, particle or composition, e.g., a treating a proliferative disorder, e.g., a cancer, in a subject, CDP-camptothecin or camptothecin derivative conjugate, e.g., a human Subject. The method comprises: particle or composition, e.g., a CDP-camptothecin or camp 0198 providing a subject who has a proliferative disorder, tothecin derivative conjugate, particle or composition e.g., Cancer, described herein, e.g., CRLX101, to said subject. 0199 administering a polysaccharide to said subject; and 0210. In one embodiment, the agent which ameliorates 0200 administering a composition that comprises a CDP bladder toxicity associated with therapy, e.g., the agent which topoisomerase inhibitor conjugate, particle or composition, increases urinary excretion and/or neutralizes one or more e.g., a CDP-topoisomerase I or II inhibitor conjugate, particle urinary metabolite, is administered prior to, concurrently or composition, e.g., a CDP-camptothecin or camptothecin with and/or after administration with the camptothecin or derivative conjugate, particle or composition, e.g., a CDP camptothecin derivative. camptothecin derivative conjugate, particle or composition 0211. In one embodiment, the agent which ameliorates described herein, e.g., CRLX101, to said subject. bladder toxicity associated with therapy is saline, e.g., intra 0201 In one embodiment, the polysaccharide is a linear, venous saline, D5 half normal saline or D5 water. In one branched or cyclic polysaccharide. In one embodiment, the embodiment, the agent which increases urinary excretion polysaccharide is a linear polysaccharide that includes glu and/or neutralizes one or more urinary metabolite is 2-mer cose molecules. In one embodiment, the polysaccharide is captoethane sulfonate sodium (MESNA). In one embodi dextran, a cyclodextrin or a cyclodextrin derivative, e.g., an ment, the agent which ameliorates bladder toxicity associated Cl-, 3- and/or Y-cyclodextrin, e.g., CDP. with therapy is 2-mercaptoethane sulfonate sodium (ME SNA) and the MESNA is administered intravenously at a 0202 In one embodiment, the polysaccharide is adminis dose of about 10%. 20%, 30% the dose of the camptothecin or tered prior to, currently with or after administration of the camptothecin derivative and/or the MESNA is administered composition. In one embodiment, the polysaccharide is orally at a dose of about 20%, 30%, 40%, 50% the dose of the administered at a dose of 100 mg to 10 g. camptothecin or camptothecin derivative. 0203. In an embodiment, the conjugate includes a topoi 0212. In an embodiment, the conjugate includes a topoi somerase I inhibitor and/or a topoisomerase II inhibitor. In an somerase I inhibitor and/or a topoisomerase II inhibitor. In an embodiment, the conjugate includes atopoisomerase I inhibi embodiment, the conjugate includes atopoisomerase I inhibi tor or combination of topoisomerase I inhibitors, e.g., camp tor or combination of topoisomerase I inhibitors, e.g., camp tothecin, irinotecan, SN-38, topotecan, lamellarin D and tothecin, irinotecan, SN-38, topotecan, lamellarin D and derivatives thereof. In an embodiment, the conjugate includes derivatives thereof. In an embodiment, the conjugate includes a topoisomerase II inhibitor or a combination of topoi a topoisomerase II inhibitor or a combination of topoi Somerase II inhibitors, e.g., eptoposide, tenoposide, doxoru Somerase II inhibitors, e.g., eptoposide, tenoposide, doxoru bicin and derivatives thereof. In one embodiment, the conju bicin and derivatives thereof. In one embodiment, the conju gate includes a combination of one or more topoisomerase I gate includes a combination of one or more topoisomerase I inhibitors and one or more topoisomerase II inhibitors. In an inhibitors and one or more topoisomerase II inhibitors. In an embodiment, the CDP-topoisomerase inhibitor conjugate is a embodiment, the CDP-topoisomerase inhibitor conjugate is a CDP-camptothecin or camptothecin derivate conjugate, e.g., CDP-camptothecin or camptothecin derivate conjugate, e.g., a CDP-camptothecin or camptothecin derivative conjugate a CDP-camptothecin or camptothecin derivative conjugate described herein, e.g., CRLX101. described herein, e.g., CRLX101. 0204. In one embodiment, the proliferative disorder is 0213. In one embodiment, the proliferative disorder is cancer, e.g., a cancer described herein. cancer, e.g., a cancer described herein. 0205. In one embodiment, the CDP-topoisomerase inhibi 0214. In one embodiment, the CDP-topoisomerase inhibi tor conjugate, particle or composition is administered incom tor conjugate, particle or composition, e.g., a CDP-topoi bination with one or more additional chemotherapeutic agent, Somerase inhibitor conjugate, particle or composition, e.g., as described herein. In one embodiment, the CDP-topoi described herein, e.g., a CDP-camptothecin or camptothecin Somerase inhibitor conjugate, particle or composition is derivative conjugate, particle or composition, e.g., a CDP administered at a dose and/or dosing schedule described camptothecin or camptothecin derivative conjugate, particle herein. In one embodiment, the Subject is administered more or composition described herein, is administered in combi than one dose of the CDP-topoisomerase inhibitor conjugate, nation with one or more additional chemotherapeutic agent, particle or composition, e.g., as described herein, and the e.g., as described herein. In one embodiment, the CDP-topoi polysaccharide is administered prior to, currently with, or Somerase inhibitor conjugate, particle or composition is after one or more dose of the CDP-topoisomerase inhibitor administered at a dose and/or dosing schedule described conjugate, particle or composition. herein. In one embodiment, the Subject is administered more 0206. In one aspect, the disclosure features, a method of than one dose of the CDP-topoisomerase inhibitor conjugate, treating a proliferative disorder, e.g., a cancer, in a subject, particle or composition, e.g., as described herein, and the e.g., a human Subject. The method comprises: agent which ameliorates bladder toxicity associated with 0207 providing a subject who has a proliferative disorder, therapy is administered prior to one or more dose of the e.g., Cancer, CDP-topoisomerase inhibitor conjugate, particle or compo 0208 administering an agent which ameliorates bladder sition. toxicity associated with therapy, e.g., an agent which 0215. In one embodiment, the CDP-topoisomerase inhibi increases urinary excretion and/or neutralizes one or more tor conjugate, particle or composition is further administered urinary metabolite; and in combination with one or more of the agents described US 2011/O160 159 A1 Jun. 30, 2011 19 herein. For example, the CDP-topoisomerase inhibitor con gate includes a combination of one or more topoisomerase I jugate, particle or composition can be administered in com inhibitors and one or more topoisomerase II inhibitors. In an bination with an agent which reduces or inhibits one or more embodiment, the CDP-topoisomerase inhibitor conjugate is a symptom of hypersensitivity. CDP-camptothecin or camptothecin derivate conjugate, e.g., 0216. In one embodiment, the method includes selecting a a CDP-camptothecin or camptothecin derivative conjugate Subject who has a proliferative disorder, e.g., cancer, and has described herein, e.g., CRLX101. experienced cystitis, e.g., has experienced cystitis as a result 0223) In one embodiment, the proliferative disorder is of a previous chemotherapeutic treatment, for administration cancer, e.g., a cancer described herein. of an agent which ameliorates bladder toxicity associated 0224. In one embodiment, the CDP-topoisomerase inhibi with therapy and a CDP-topoisomerase inhibitor conjugate, tor conjugate, particle or composition is administered in com particle or composition, e.g., a CDP-camptothecin or camp bination with one or more additional chemotherapeutic agent, tothecin derivative conjugate, particle or composition, e.g., a e.g., as described herein. In one embodiment, the CDP-topoi CDP-camptothecin or camptothecin derivative conjugate, Somerase inhibitor conjugate, particle or composition is particle or composition described herein, e.g., CRLX101. administered at a dose and/or dosing schedule described 0217. In one aspect, the disclosure features, a method of herein. In one embodiment, the Subject is administered more treating a proliferative disorder, e.g., a cancer, in a subject, than one dose of the CDP-topoisomerase inhibitor conjugate, e.g., a human Subject. The method comprises: particle or composition, e.g., as described herein, and the 0218 providing a subject who has a proliferative disorder, agent which reduces or inhibits one or more symptom of e.g., cancer, and has been administered an agent which hypersensitivity is administered prior to each dose of the reduces or inhibits one or more symptom of hyperSensitivity; CDP-topoisomerase inhibitor conjugate, particle or compo and sition. 0219 administering a composition that comprises a CDP 0225. In one embodiment, the method includes selecting a topoisomerase inhibitor conjugate, particle or composition, Subject who has a proliferative disorder, e.g., cancer, and has e.g., a CDP-camptothecin or camptothecin derivative conju experienced one or more symptom of hypersensitivity, e.g., gate, particle or composition, e.g., a CDP-camptothecin or has experienced one or more symptom of hyperSensitivity to camptothecin derivative conjugate, particle or composition a previous chemotherapeutic treatment, for administration of described herein, e.g., CRLX101, to said subject. an agent which reduces or inhibits one or more symptom of 0220. In one embodiment, the method further comprises hypersensitivity and a CDP-topoisomerase inhibitor conju administering the agent which reduces or inhibits one or more gate, particle or composition, e.g., a CDP-camptothecin or symptom of hypersensitivity to the Subject. camptothecin derivative conjugate, particle or composition, 0221. In one embodiment, the agent which reduces or e.g., a CDP-camptothecin or camptothecin derivative conju inhibits one or more symptoms of hypersensitivity can be one gate, particle or composition described herein, e.g., or more of a (e.g., dexamethasone), an antihis CRLX101. Symptoms of hypersensitivity include: injection tamine (e.g., diphenhydramine), an and an H2 site reaction, dyspnea, hypotension, angioedema, urticaria, antagonist (e.g., ranitidine or famotidine). In one embodi bronchospasm and erythema. ment, the agent is a corticosteroid (e.g., dexamethasone) and 0226. In yet another aspect, the disclosure features a the corticosteroid is administered at 5, 10, 15, 20, 25 or 30 mg. method of treating a Subject, e.g., a human Subject, with a In one embodiment, the corticosteroid is administered about proliferative disorder, e.g., cancer, comprising: 12, 11, 10,9,8,7,6, 5, 4, and/or 3 hours before administration 0227 selecting a subject who has a proliferative disorder, of the CDP-topoisomerase inhibitor conjugate, particle or e.g., cancer, that has increased KRAS and/or ST expression composition, or the corticosteroid is administered intrave levels, e.g., as compared to a reference standard and/or a nously about 40, 30, 20 minutes before the CDP-topoi mutation in a KRAS gene and/or ST gene; and Somerase inhibitor conjugate, particle or composition. In one 0228 administering a CDP-topoisomerase inhibitor con embodiment, the agent is an antihistamine (e.g., diphenhy jugate, particle or composition, e.g., a CDP-camptothecin or dramine) and the antihistamine is administered at 15, 20, 25, camptothecin derivative conjugate, particle or composition, 30,35, 40, 45,50, 55,60, 65 or 70 mg. In one embodiment, the e.g., a CDP-camptothecin or camptothecin derivative conju antihistamine is administered intravenously about 40, 30, 20, gate, particle or composition described herein, e.g., 10 minutes before the CDP-topoisomerase inhibitor conju CRLX101, to the subject in an amount effective to treat the gate, particle or composition. In one embodiment, the agentis cancer, to thereby treat the cancer. an (e.g., ranitidine or famotidine) and the H2 0229. In an embodiment, the conjugate includes a topoi antagonist is administered at 10, 15, 20, 25, 30, 35, 40, 45, 50. somerase I inhibitor and/or a topoisomerase II inhibitor. In an 55, 60, 65 or 70 mg. In one embodiment the H2 antagonist is embodiment, the conjugate includes atopoisomerase I inhibi administered intravenously about 70, 60, 50, 40, 30, 20, 10 tor or combination of topoisomerase I inhibitors, e.g., camp minutes before the CDP-topoisomerase inhibitor conjugate, tothecin, irinotecan, SN-38, topotecan, lamellarin D and particle or composition. derivatives thereof. In an embodiment, the conjugate includes 0222. In an embodiment, the conjugate includes a topoi a topoisomerase II inhibitor or a combination of topoi somerase I inhibitor and/or a topoisomerase II inhibitor. In an Somerase II inhibitors, e.g., eptoposide, tenoposide, doxoru embodiment, the conjugate includes atopoisomerase I inhibi bicin and derivatives thereof. In one embodiment, the conju tor or combination of topoisomerase I inhibitors, e.g., camp gate includes a combination of one or more topoisomerase I tothecin, irinotecan, SN-38, topotecan, lamellarin D and inhibitors and one or more topoisomerase II inhibitors. In an derivatives thereof. In an embodiment, the conjugate includes embodiment, the CDP-topoisomerase inhibitor conjugate is a a topoisomerase II inhibitor or a combination of topoi CDP-camptothecin or camptothecin derivate conjugate, e.g., Somerase II inhibitors, e.g., eptoposide, tenoposide, doxoru a CDP-camptothecin or camptothecin derivative conjugate bicin and derivatives thereof. In one embodiment, the conju described herein, e.g., CRLX101. US 2011/O160 159 A1 Jun. 30, 2011 20

0230. In one embodiment, the subject has increased topoisomerase inhibitor conjugate including an inclusion KRAS and/or ST expression levels, e.g., as compared to a complex forms a nanoparticle. The nanoparticle ranges in reference standard, and/or has a mutation in a KRAS and/or size from 10 to 300 nm in diameter, e.g., 20 to 280, 30 to 250, ST gene. In one embodiment, the Subject has a mutation at 30 to 200, 20 to 150, 30 to 100, 20 to 80, 30 to 70, 30 to 60 or one or more of codon 12 of the KRAS gene (e.g., a G to T transversion), codon 13 of the KRAS gene, codon 61 of the 30 to 50 nm diameter. In one embodiment, the nanoparticle is KRAS gene. In one embodiment, the Subject has lung cancer 30 to 60 nm in diameter. In one embodiment, the composition (e.g., Small cell lung cancer and/or non-Small cell lung can comprises a population or a plurality of nanoparticles with an cer), pancreatic cancer or colorectal cancer. average diameter from 10 to 300 nm, e.g., 20 to 280, 30 to 0231. In one embodiment, the cancer is a cancer described 250, 30 to 200, 20 to 150, 30 to 100, 20 to 80, 30 to 70, 30 to herein. 60 or 30 to 50 nm. In one embodiment, the average nanopar 0232. In one embodiment, the CDP-topoisomerase inhibi ticle diameter is from 30 to 60 nm. In one embodiment, the tor conjugate, particle or composition is administered incom Surface charge of the molecule is neutral, or slightly negative. bination with one or more additional chemotherapeutic agent, In some embodiments, the Zeta potential of the particle Sur e.g., as described herein. In one embodiment, the CDP-topoi face is from about -80 mV to about 50 mV, about -20 mV to Somerase inhibitor conjugate, particle or composition is about 20 mV, about -20 mV to about -10 mV, or about -10 administered at a dose and/or dosing schedule described mV to about 0. herein. 0240. In one embodiment, the topoisomerase inhibitor 0233. In one embodiment, the CDP-topoisomerase inhibi (e.g., a camptothecin derivative, e.g., a camptothecin deriva tor conjugate, particle or composition is administered incom tive described herein), conjugated to the CDP is more soluble bination with one or more of the agents described herein. For when conjugated to the CDP than when not conjugated to the example, the CDP-topoisomerase inhibitor conjugate, par CDP. ticle or composition can be administered in combination with an agent which reduces or inhibits one or more symptom of 0241. In one embodiment, the composition comprises a hypersensitivity and/or an agent which increases urinary population, mixture or plurality of CDP-topoisomerase excretion and/or neutralizes one or more urinary metabolite. inhibitor conjugates. In one embodiment, the population, 0234. In another aspect, the disclosure features, a unit mixture or plurality of CDP-topoisomerase inhibitor conju dosage of a CDP-topoisomerase inhibitor conjugate, particle gates comprises a plurality of different topoisomerase inhibi or composition, e.g., a CDP-camptothecin or camptothecin tors conjugated to a CDP (e.g., two different topoisomerase derivative conjugate, particle or composition, e.g., a CDP inhibitors are in the composition such that two different topoi camptothecin or camptothecin derivative conjugate, particle somerase inhibitors are attached to a single CDP; or a first or composition described herein, e.g., CRLX101. topoisomerase inhibitor is attached to a first CDP and a sec 0235. In one aspect, the disclosure features a CDP-topoi ond topoisomerase inhibitor is attached to a second CDP and Somerase inhibitor conjugate, particle or composition, e.g., a both CDP-topoisomerase inhibitor conjugates are present in CDP-camptothecin derivative conjugate, particle or compo the composition). sition, e.g., a CDP-camptothecin derivative conjugate, par ticle or composition described herein, and methods of making 0242. The details of one or more embodiments of the the CDP-topoisomerase inhibitor conjugates, particles and invention are set forth in the description below. Other fea compositions, e.g., a CDP-camptothecin derivative conju tures, objects, and advantages of the invention will be appar gate, particle or composition, e.g., a CDP-camptothecin ent from the description and the drawings, and from the derivative conjugate, particle or composition described claims. herein. 0236. In an embodiment, the conjugate includes a topoi BRIEF DESCRIPTION OF THE DRAWINGS somerase I inhibitor and/or a topoisomerase II inhibitor. In an embodiment, the conjugate includes atopoisomerase I inhibi 0243 FIGS. 1A and 1B are CT (computed tomography) tor or combination of topoisomerase I inhibitors, e.g., irino scans from a patient with metastatic pancreatic cancer pre tecan, SN-38, topotecan, lamellarin D and derivatives treatment (FIG. 1A), and after six months of treatment with thereof. In an embodiment, the conjugate includes a topoi CRLX101 (FIG. 1B). The patient received 6 mg/m somerase II inhibitor or a combination of topoisomerase II CRLX101 on Schedule Ia. inhibitors, e.g., eptoposide, tenoposide, doxorubicin and 0244 FIGS. 2A and 2B are graphs depicting pharmacoki derivatives thereof. In one embodiment, the conjugate includes a combination of one or more topoisomerase I netic and toxicokinetic analysis of CRLX101 delivered by inhibitors and one or more topoisomerase II inhibitors. intravenous administration. FIG. 2A shows mean plasma 0237. In one embodiment, CDP is not biodegradable. concentration-time profile for polymer conjugated (squares) 0238. In one embodiment, CDP is biocompatible. and unconjugated (triangles) CPT for cohort 1b-1 (12 0239. In one embodiment, the CDP-topoisomerase inhibi mg/m). FIG.2B shows average urinary excretion of polymer tor conjugate, particle or composition, e.g., a CDP-camptoth conjugated (black bars) and unconjugated CPT (white bars) ecin derivative conjugate particle or composition, e.g., a in the first 48 hours following CRLX101 administration. CDP-camptothecin derivative described herein, includes an Plasma concentrations for conjugated and unconjugated CPT inclusion complex between a topoisomerase inhibitor, e.g., a were below the limit of quantitation at 336 hrs (before the camptothecin derivative, attached or conjugated to the CDP. second dose) and therefore are not plotted in FIG. 2A. e.g., via a covalent linkage, and another molecule in the CDP. 0245 FIGS. 3A-3C depict immunohistochemistry and In one embodiment, the CDP-topoisomerase inhibitor conju topoisomerase I activity of ovarian cancer cells from a patient gate forms a nanoparticle. In one embodiment, the CDP treated with CRLX101, showing post-treatment reduction of US 2011/O160 159 A1 Jun. 30, 2011 topoisomerase I protein. FIG.3A is an immunohistochemical

stain of ascites cells collected before CRLX101 was given. FIG. 3B is an immunohistochemical stain of ascites cells collected 2 days following CRLX101 treatment. FIG. 3C is a gel depicting the results oftopoisomerase I enzymatic activity assay in whole cell lysates. 0246 FIG. 4 depicts the structure and description of an exemplary CDP-camptothecin conjugate referred to as “CRLX101 throughout this application. CRLX101 is used interchangeably with the term CRLX101 (e.g., as in Example 1).

DETAILED DESCRIPTION OF THE INVENTION 0252) wherein 0253) R' is H, OH, optionally substituted (e.g., 0247 The present invention relates to compositions of optionally substituted with NR' or OR, or SiR), or SiR: therapeutic cyclodextrin-containing polymers (CDP) or R' and R may be taken together to form an optionally designed for drug delivery of a topoisomerase inhibitor Such Substituted 5- to 8-membered ring (e.g., optionally substi tuted with NR", or OR): as camptothecin or a camptothecin derivative. In certain 0254 R is H, OH, NH, halo, nitro, optionally substituted embodiments, these cyclodextrin-containing polymers alkyl (e.g., optionally substituted with NR or OR, NR", improve drug stability and/or , and/or reduce toxic OC(=O)NR', or OC(=O)CR): ity, and/or improve efficacy of the topoisomerase inhibitor (0255 R is H, OH, NH, halo, nitro, NR", OC(=O) when used in vivo. NR, or OC(=O)CR 0248. Furthermore, by selecting from a variety of linker 0256 R is H, OH, NH, halo, CN, or NR; or Rand R' groups that link or couple CDP to a topoisomerase inhibitor taken together with the atoms to which they are attached form Such as camptothecin or a camptothecin derivative, and/or a 5- or 6-membered ring (e.g. forming a ring including targeting ligands, the rate of drug release from the polymers –OCHO or —OCHCHO ); can be attenuated for controlled delivery. The invention also (0257 each R is independently H or alkyl: or two R's, taken together with the atom to which they are attached, form relates to methods of treating Subjects with compositions a 4- to 8-membered ring (e.g., optionally containing an O or described herein. The invention further relates to methods for NR) conducting a pharmaceutical business comprising manufac 0258 R, is H or optionally substituted alkyl (e.g., option turing, licensing, or distributing kits containing or relating to ally substituted with OR or NR): the CDP-topoisomerase inhibitor conjugates, particles and (0259 R is Horalkyl; or, two R's, taken together with the compositions described herein. atom to which they are attached, form a 4- to 8-membered 0249 More generally, the present invention provides ring; and water-soluble, biocompatible polymer conjugates compris 0260 n=0 or 1. ing a water-soluble, biocompatible polymer covalently 0261. In some embodiments, the camptothecin or camp attached to the topoisomerase inhibitor through attachments tothecin derivative is the compound as provided below. that are cleaved under biological conditions to release the topoisomerase inhibitor. 0250 Polymeric conjugates featured in the methods described herein may be useful to improve solubility and/or stability of a bioactive/therapeutic agent, such as camptoth ecin, reduce drug-drug interactions, reduce interactions with blood elements including plasma , reduce or elimi nate immunogenicity, protect the agent from , modulate drug-release kinetics, improve circulation time, improve drug half-life (e.g., in the serum, or in selected tis Sues, such as tumors), attenuate toxicity, improve efficacy, normalize drug metabolism across Subjects of different spe cies, ethnicities, and/or races, and/or provide for targeted 0262. In one embodiment, R. R. RandR of the camp delivery into specific cells or tissues. tothecin derivative are each H, and n is 0. 0251. In preferred embodiments, the topoisomerase 0263. In one embodiment, R. R. RandR of the camp inhibitor in the CDP-topoisomerase inhibitor conjugate, par tothecin derivative are each H, and n is 1. ticle or composition is camptothecin or a camptothecin 0264. In one embodiment, R' of the camptothecin deriva derivative. The term “camptothecin derivative', as used tive is H, R is —CHN(CH), R is -OH, R is H; and n is herein, includes camptothecin analogues and metabolites of O camptothecin. For example, camptothecin derivatives can 0265. In one embodiment, R' of the camptothecin deriva have the following structure: tive is —CHCH R is H, US 2011/O160 159 A1 Jun. 30, 2011 22

0266 R is:

ry s

R is H, and n is O. In one embodiment, R is: 0267. In one embodiment, R' of the camptothecin deriva tive is CHCH R is H, R is -OH, R is H, and n is 0. 0268. In one embodiment, R' of the camptothecin deriva tive is tert-butyldimethylsilyl, R is H, R is –OH and R is H, and n is 0. 0269. In one embodiment, R' of the camptothecin deriva tive is tert-butyldimethylsilyl, R is hydrogen, Ris-OH and R" is hydrogen, and n is 1. and R is hydrogen. (0270. In one embodiment, R' of the camptothecin deriva 0275. In one embodiment, the camptothecin derivative has tive is tert-butyldimethylsilyl, R, R and Rare each H, and the following formula: n is 0. 0271. In one embodiment, R' of the camptothecin deriva tive is tert-butyldimethylsilyl, R. RandR are each H, and n is 1. (0272. In one embodiment, R' of the camptothecin deriva tive is —CHCHSi(CH), and R, R and Rare each H. (0273. In one embodiment, R' and R of the camptothecin derivative are taken together with the to which they are attached to form an optionally Substituted ring. In one embodiment, R' and R of the camptothecin derivative are taken together with the carbons to which they are attached to form a substituted 6-membered ring. In one embodiment, the camptothecin derivative has the following formula: In one embodiment, R is:

and R is hydrogen. (0276. In one embodiment, R is:

In one embodiment, R is methyl and R is fluoro. (0274) In one embodiment, R and R are taken together with the carbons to which they are attached to forman option ally substituted ring. In one embodiment, Rand Rare taken together with the carbons to which they are attached to form R’ is H, R is methyl, R is chloro; and n is 1. a 6-membered heterocyclic ring. In one embodiment, the (0277. In one embodiment, R is CH=NOC(CH), R, camptothecin derivative has the following formula: RandR are each H, and n is 0. US 2011/O160 159 A1 Jun. 30, 2011

0278. In one embodiment, R' is —CHCH-NHCH(CH) 0287. Accordingly, in one embodiment the CDP-topoi R. Rand Rare each H; and n is 0. Somerase inhibitor conjugate is represented by Formula I: (0279. In one embodiment, R' and Rare H, Rand Rare fluoro, and n is 1. 0280. In one embodiment, each of R', R, and R is H. R. is NH, and n is 0. (I) (0281. In one embodiment, each of R', R, and R is H. R. is NO, and n is 0. 0282. An “effective amount’ or “an amount effective' refers to an amount of the CDP-topoisomerase inhibitor con jugate, particle or composition which is effective, upon single ormultiple dose administrations to a subject, intreating a cell, or curing, alleviating, relieving or improving a symptom of a disorder. An effective amount of the conjugate, particle or composition may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicita desired response in the individual. An 0288 wherein effective amount is also one in which any toxic or detrimental effects of the conjugate, particle or composition is out 0289 P represents a linear or branched polymer chain; weighed by the therapeutically beneficial effects. 0290 CD represents a cyclic moiety such as a cyclodextrin 0283 As used herein, the term “subject' is intended to moiety; include human and non-human animals. Exemplary human 0291 L. L and L, independently for each occurrence, Subjects include a human patient having a disorder, e.g., a may be absent or represent a linker group; disorder described herein, or a normal subject. The term “non-human animals' includes all vertebrates, e.g., non 0292 D, independently for each occurrence, represents a mammals (Such as chickens, amphibians, reptiles) and mam topoisomerase inhibitor or a prodrug thereof (e.g., a camp mals. Such as non-human primates, domesticated and/or agri tothecin or camptothecin derivative); culturally useful animals, e.g., sheep, dog, cat, cow, pig, etc. 0293 T independently for each occurrence, represents a 0284 As used herein, the term “treat' or “treating a sub targeting ligand or precursor thereof; ject having a disorder refers to Subjecting the Subject to a 0294 a.m., and V, independently for each occurrence, rep regimen, e.g., the administration of a CDP-topoisomerase resent integers in the range of 1 to 10 (preferably 1 to 8, 1 to inhibitor conjugate, particle or composition, Such that at least one symptom of the disorder is cured, healed, alleviated, 5, or even 1 to 3): relieved, altered, remedied, ameliorated, or improved. Treat 0295 n and w, independently for each occurrence, repre ing includes administering an amount effective to alleviate, sent an integer in the range of 0 to about 30,000 (preferably relieve, alter, remedy, ameliorate, improve or affect the dis <25,000, <20,000, <15,000, <10,000, <5,000, <1,000, <500, order or the symptoms of the disorder. The treatment may <100, <50. <25, <10, or even <5); and inhibit deterioration or worsening of a symptom of a disorder. 0296 b represents an integer in the range of 1 to about 0285 An amount of a CDP-topoisomerase inhibitor con 30,000 (preferably <25,000, <20,000, <15,000, <10,000, jugate, particle or composition effective to prevent a disorder, or “a prophylactically effective amount of the conjugate, <5,000, <1,000, <500, <100,<50. <25, <10, or even <5), particle or composition as used in the context of the admin 0297 wherein either P comprises cyclodextrin moieties or istration of an agent to a Subject, refers to Subjecting the n is at least 1. Subject to a regimen, e.g., the administration of a CDP-topoi 0298. In some embodiments, one or more of the topoi Somerase inhibitor conjugate, particle or composition Such somerase inhibitor moieties in the CDP-topoisomerase that the onset of at least one symptom of the disorder is inhibitor conjugate can be replaced with another therapeutic delayed as compared to what would be seen in the absence of agent, e.g., another anticancer agent or anti-inflammatory the regimen. agent. Examples of other anticancer agents are described CDP-Topoisomerase Inhibitor Conjugates, Particles and herein. Examples of anti-inflammatory agents include a ste Compositions roid, e.g., prednisone, and a NSAID. 0299. In certain embodiments, P contains a plurality of 0286. Described herein are cyclodextrin containing poly cyclodextrin moieties within the polymer chain as opposed to mer (“CDP)-topoisomerase inhibitor conjugates, wherein the cyclodextrin moieties being grafted on to pendant groups one or more topoisomerase inhibitors are covalently attached to the CDP (e.g., either directly or through a linker). The off of the polymeric chain. Thus, in certain embodiments, the CDP-topoisomerase inhibitor conjugates include linear or polymer chain of formula I further comprises n' units of U. branched cyclodextrin-containing polymers and polymers wherein n' represents an integer in the range of 1 to about grafted with cyclodextrin. Exemplary cyclodextrin-contain 30,000, e.g., from 4-100, 4-50, 4-25, 4-15, 6-100, 6-50, 6-25, ing polymers that may be modified as described herein are and 6-15 (preferably <25,000, <20,000, <15,000, <10,000, taught in U.S. Pat. Nos. 7,270.808, 6,509,323, 7,091,192, <5,000, <1,000, <500, <100, <50, <25, <20, <15, <10, or 6,884,789, U.S. Publication Nos. 20040087024, even <5); and U is represented by one of the general formulae 2004O109888 and 2007OO25952. below: US 2011/O160 159 A1 Jun. 30, 2011 24

ypropyl)methacrylamide (HPMA), dextran and hydroxy ethyl starch (HES), and have optional pendant groups for grafting therapeutic agents, targeting ligands and/or cyclo dextrin moieties. In certain embodiments, the polymer may be biodegradable Such as poly(lactic acid), poly(glycolic acid), poly(alkyl 2-cyanoacrylates), polyanhydrides, and polyorthoesters, or bioerodible such as polylactide-glycolide copolymers, and derivatives thereof, non-peptide polyami noacids, polyiminocarbonates, polyalpha-amino acids, poly alkyl-cyano-acrylate, polyphosphaZenes or acyloxymethyl poly aspartate and polyglutamate copolymers and mixtures thereof. 0311. In another embodiment the CDP-topoisomerase p inhibitor conjugate is represented by Formula II: L5 L4 CD L7 (II) (D), (D), (T)),

0300 wherein 0301 CD represents a cyclic moiety, such as a cyclodex L10 L9 trin moiety, or derivative thereof; (CD) (D), 0302 La Ls. Le, and L7, independently for each occur pi g rence, may be absent or represent a linker group; 0303 D and D', independently for each occurrence, rep 0312 wherein resent the same or different topoisomerase inhibitor or pro 0313 Prepresents a monomer unit of a polymer that com drug forms thereof (e.g., a camptothecin or camptothecin prises cyclodextrin moieties; derivative); 0304 T and T', independently for each occurrence, repre 0314 T independently for each occurrence, represents a sent the same or different targeting ligand or precursor targeting ligand or a precursor thereof; thereof 0315 L. L., Ls, Lo, and Lo, independently for each 0305 fandy, independently for each occurrence, repre occurrence, may be absent or represent a linker group; sent an integer in the range of 1 and 10; and 0316 CD, independently for each occurrence, represents a 0306 g and Z, independently for each occurrence, repre cyclodextrin moiety or a derivative thereof; sent an integer in the range of 0 and 10. 0317 D, independently for each occurrence, represents a 0307 Preferably the polymer has a plurality of D or D' topoisomerase inhibitor or a prodrug form thereof (e.g., a moieties. In some embodiments, at least 50% of the U units camptothecin or camptothecin derivative); have at least one D or D'. In some embodiments, one or more 0318 m, independently for each occurrence, represents an of the topoisomerase inhibitor moieties in the CDP-topoi integer in the range of 1 to 10 (preferably 1 to 8, 1 to 5, or even Somerase conjugate can be replaced with another therapeutic 1 to 3): agent, e.g., another anticancer agent or anti-inflammatory 0319 o represents an integer in the range of 1 to about agent. 30,000 (preferably <25,000, <20,000, <15,000, <10,000, 0308. In preferred embodiments, L and L, represent linker groups. <5,000, <1,000, <500,<100,<50, <25, <10, or even <5); and 0309 The CDP may include a polycation, polyanion, or 0320 p, n, and q, independently for each occurrence, rep non-ionic polymer. A polycationic or polyanionic polymer resent an integer in the range of 0 to 10 (preferably 0 to 8, 0 to has at least one site that bears a positive or negative charge, 5, 0 to 3, or even 0 to about 2), respectively. In certain Such embodiments, at least one of the 0321 wherein CD and D are preferably each present at linker moiety and the cyclic moiety comprises such a charged least 1 location (preferably at least 5, 10, 25, or even 50 or 100 site, so that every occurrence of that moiety includes a locations) in the compound. charged site. In some embodiments, the CDP is biocompat 0322. In some embodiments, one or more of the topoi ible. somerase inhibitor moieties in the CDP-topoisomerase 0310. In certain embodiments, the CDP may include inhibitor conjugate can be replaced with another therapeutic polysaccharides, and other non-protein biocompatible poly agent, e.g., another anticancer agent or anti-inflammatory mers, and combinations thereof, that contain at least one agent. Examples of an anticancer agent are described herein. terminal hydroxyl group, Such as polyvinylpyrrollidone, poly Examples of anti-inflammatory agents include a steroid, e.g., (Oxyethylene)glycol (PEG), poly Succinic anhydride, polyse prednisone, or a NSAID. bacic acid, PEG-, polyglutamate, polyethylen 0323. In another embodiment the CDP-topoisomerase imine, maleic anhydride divinylether (DIVMA), cellulose, inhibitor conjugate is represented either of the formulae pullulans, inulin, polyvinyl alcohol (PVA), N-(2-hydrox below: US 2011/O160 159 A1 Jun. 30, 2011 25

(e.g., 18-crown-6,15-crown-5,12-crown-4, etc.), cyclic oli gopeptides (e.g., comprising from 5 to 10 amino acid resi (T), dues), cryptands or cryptates (e.g., cryptand 2.2.2, cryptand-2,1,1, and complexes thereof), calixarenes, or cav L5 2 itands, or any combination thereof. Preferably, the cyclic structure is (or is modified to be) water-soluble. In certain embodiments, e.g., for the preparation of a linear polymer, the L7 O cyclic structure is selected Such that under polymerization conditions, exactly two moieties of each cyclic structure are (D), (T), reactive with the linker moieties, such that the resulting poly 8 L6 2 mer comprises (or consists essentially of) an alternating series of cyclic moieties and linker moieties, such as at least (D), 8 four of each type of moiety. Suitable difunctionalized cyclic moieties include many that are commercially available and/or amenable to preparation using published protocols. In certain embodiments, conjugates are soluble in water to a concentra tion of at least 0.1 g/mL, preferably at least 0.25 g/mL. 0336. Thus, in certain embodiments, the invention relates to novel compositions of therapeutic cyclodextrin-containing polymeric compounds designed for drug delivery of a topoi somerase inhibitor. In certain embodiments, these CDPs improve drug stability and/or solubility, and/or reduce toxic ity, and/or improve efficacy of the topoisomerase inhibitor when used in vivo. Furthermore, by selecting from a variety of linker groups, and/or targeting ligands, the rate of topoi 0324 wherein somerase inhibitor release from the CDP can be attenuated 0325 CD represents a cyclic moiety, such as a cyclodex for controlled delivery. trin moiety, or derivative thereof; 0337. In certain embodiments, the CDP comprises a linear 0326 La Ls, L and L, independently for each occur cyclodextrin-containing polymer, e.g., the polymer backbone rence, may be absent or represent a linker group; includes cyclodextrin moieties. For example, the polymer 0327 D and D', independently for each occurrence, rep may be a water-soluble, linear cyclodextrin polymer pro resent the same or different topoisomerase inhibitor or pro duced by providing at least one cyclodextrin derivative modi drug thereof (e.g., a camptothecin or camptothecin deriva fied to bear one reactive site at each of exactly two positions, tive); and reacting the cyclodextrin derivative with a linker having 0328 T and T', independently for each occurrence, repre exactly two reactive moieties capable of forming a covalent sent the same or different targeting ligand or precursor bond with the reactive sites under polymerization conditions thereof that promote reaction of the reactive sites with the reactive 0329 fandy, independently for each occurrence, repre moieties to form covalent bonds between the linker and the sent an integer in the range of 1 and 10 (preferably 1 to 8, 1 to cyclodextrin derivative, whereby a linear polymer compris 5, or even 1 to 3): ing alternating units of cyclodextrin derivatives and linkers is 0330 g and Z, independently for each occurrence, repre produced. Alternatively the polymer may be a water-soluble, sent an integer in the range of 0 and 10 (preferably 0 to 8, 0 to linear cyclodextrin polymer having a linear polymer back 5, 0 to 3, or even 0 to about 2); and bone, which polymer comprises a plurality of substituted or 0331 h represents an integer in the range of 1 and 30,000, unsubstituted cyclodextrin moieties and linker moieties in the e.g., from 4-100, 4-50, 4-25, 4-15, 6-100, 6-50, 6-25, and 6-15 linear polymer backbone, wherein each of the cyclodextrin (preferably <25,000, <20,000, <15,000, <10,000, <5,000, moieties, other than a cyclodextrin moiety at the terminus of <1,000, <500, <100, <50. <25, <20, <15, <10, or even <5), a polymer chain, is attached to two of said linker moieties, 0332 wherein at least one occurrence (and preferably at each linker moiety covalently linking two cyclodextrin moi least 5, 10, or even at least 20, 50, or 100 occurrences) ofg eties. In yet another embodiment, the polymer is a water represents an integer greater than 0. soluble, linear cyclodextrin polymer comprising a plurality of 0333 Preferably the polymer has a plurality of D or D' cyclodextrin moieties covalently linked together by a plural moieties. In some embodiments, at least 50% of the polymer ity of linker moieties, wherein each cyclodextrin moiety, repeating units have at least one D or D'. In some embodi other than a cyclodextrin moiety at the terminus of a polymer ments, one or more of the topoisomerase inhibitor moieties in chain, is attached to two linker moieties to form a linear the CDP-topoisomerase inhibitor conjugate can be replaced cyclodextrin polymer. with another therapeutic agent, e.g., another anticancer agent 0338. In some embodiments, the CDP-topoisomerase or anti-inflammatory agent. inhibitor conjugate comprises a water soluble linear polymer 0334. In preferred embodiments, La and L, represent conjugate comprising: cyclodextrin moieties; comonomers linker groups. which do not contain cyclodextrin moieties (comonomers); 0335. In certain such embodiments, the CDP comprises and a plurality of topoisomerase inhibitor; wherein the CDP cyclic moieties alternating with linker moieties that connect topoisomerase inhibitor conjugate comprises at least four, the cyclic , e.g., into linear or branched polymers, five six, seven, eight, etc., cyclodextrin moieties and at least preferably linear polymers. The cyclic moieties may be any four, five six, seven, eight, etc., comonomers. In some Suitable cyclic structures, such as cyclodextrins, crown ethers embodiments, the topoisomerase inhibitor is a topoisomerase US 2011/O160 159 A1 Jun. 30, 2011 26 inhibitor described herein, for example, the topoisomerase hours, 3 hours, 5 hours, 6 hours, 8 hours, 10 hours, 15 hours, inhibitor is a camptothecin or camptothecin derivative 20 hours, 1 day, 2 days, 3 days, 4 days, 7 days, 10 days, 14 described herein. The topoisomerase inhibitor can be days, 17 days, 20 days, 24 days, 27 days up to a month. In attached to the CDP via a functional group such as a hydroxyl some embodiments, upon administration of the CDP-topoi group, or where appropriate, an amino group. Somerase inhibitor conjugate to a subject, the rate of topoi 0339. In some embodiments, one or more of the topoi Somerase inhibitor release is dependent primarily upon the somerase inhibitor moieties in the CDP-topoisomerase rate of hydrolysis as opposed to enzymatic cleavage. inhibitor conjugate can be replaced with another therapeutic 0345. In some embodiments, the CDP-topoisomerase agent, e.g., another anticancer agent or anti-inflammatory inhibitor conjugate has a molecular weight of 10,000-500, agent. 000. In some embodiments, the cyclodextrin moieties make 0340. In some embodiments, the least four cyclodextrin up at least about 2%. 5%, 10%, 20%, 30%, 50% or 80% of the moieties and at least four comonomers alternate in the CDP CDP-topoisomerase inhibitor conjugate by weight. topoisomerase inhibitor conjugate. In some embodiments, 0346. In some embodiments, the CDP-topoisomerase the topoisomerase inhibitors are cleaved from said CDP inhibitor conjugate is made by a method comprising provid topoisomerase inhibitor conjugate under biological condi ing cyclodextrin moiety precursors modified to bear one reac tions to release the topoisomerase inhibitor. In some embodi tive site at each of exactly two positions, and reacting the ments, the cyclodextrin moieties comprise linkers to which cyclodextrin moiety precursors with comonomer precursors topoisomerase inhibitors are linked In some embodiments, having exactly two reactive moieties capable of forming a the topoisomerase inhibitors are attached via linkers. covalent bond with the reactive sites under polymerization 0341. In some embodiments, the comonomer comprises conditions that promote reaction of the reactive sites with the residues of at least two functional groups through which reactive moieties to form covalent bonds between the reaction and linkage of the cyclodextrin monomers was comonomers and the cyclodextrin moieties, whereby a CDP achieved. In some embodiments, the functional groups, comprising alternating units of a cyclodextrin moiety and a which may be the same or different, terminal or internal, of comonomer is produced. In some embodiments, the cyclo each comonomer comprise an amino, acid, imidazole, dextrin moiety precursors are in a composition, the compo hydroxyl, thio, acyl halide, —HC=CH-, -C=C-group, sition being Substantially free of cyclodextrin moieties having or derivative thereof. In some embodiments, the two func other than two positions modified to bear a reactive site (e.g., tional groups are the same and are located at termini of the cyclodextrin moieties having 1, 3, 4, 5, 6, or 7 positions comonomer precursor. In Some embodiments, a comonomer modified to bear a reactive site). contains one or more pendant groups with at least one func 0347 In some embodiments, a comonomer of the CDP tional group through which reaction and thus linkage of a topoisomerase inhibitor conjugate comprises a moiety topoisomerase inhibitor was achieved. In some embodi selected from the group consisting of an alkylene chain, ments, the functional groups, which may be the same or poly Succinic anhydride, poly-L-glutamic acid, poly(ethyl different, terminal or internal, of each comonomer pendant eneimine), an oligosaccharide, and an amino acid chain. In group comprise an amino, acid, imidazole, hydroxyl, thiol, some embodiments, a CDP-topoisomerase inhibitor conju acyl halide, ethylene, ethyne group, or derivative thereof. In gate comonomer comprises a polyethylene glycol chain. In Some embodiments, the pendant group is a Substituted or Some embodiments, a comonomer comprises a moiety unsubstituted branched, cyclic or straight chain C1-C10 selected from: polyglycolic acid and polylactic acid chain. In alkyl, orarylalkyl optionally containing one or more heteroa Some embodiments, a comonomer comprises a hydrocarby toms within the chain or ring. In some embodiments, the lene group wherein one or more methylene groups is option cyclodextrin moiety comprises an alpha, beta, or gamma ally replaced by a group Y (provided that none of theY groups cyclodextrin moiety. In some embodiments, the topoi are adjacent to each other), wherein each Y, independently for somerase inhibitor is at least 5%, 10%, 15%, 20%, 25%, 30%, each occurrence, is selected from, Substituted or unsubsti or 35% by weight of CDP-topoisomerase inhibitor conjugate. tuted aryl, heteroaryl, cycloalkyl, heterocycloalkyl, or 0342. In some embodiments, the comonomer comprises —O—, C(=X) (wherein X is NRO or S), —OC(O)—, polyethylene glycol of molecular weight 3,400 Da, the cyclo —C(=O)C), NRCO , —C(O)NR— —S(O), dextrin moiety comprises beta-cyclodextrin, the theoretical (wherein n is 0, 1, or 2), —OC(O)—NR, —NR—C(O) maximum loading of the topoisomerase inhibitor on the NR—, NR1-C(NR) NR—, and —B(OR)—; and CDP-topoisomerase inhibitor conjugate is 13% by weight, R, independently for each occurrence, represents H or a and the topoisomerase inhibitor is 6-10% by weight of CDP lower alkyl. topoisomerase inhibitor conjugate. In some embodiments, 0348. In some embodiments, the CDP-topoisomerase the topoisomerase inhibitor is poorly soluble in water. In inhibitor conjugate is a polymer having attached thereto a some embodiments, the solubility of the topoisomerase plurality of D moieties of the following formula: inhibitor is <5 mg/ml at physiological pH. In some embodi ments, the topoisomerase inhibitor is a hydrophobic com pound with a log Pid-0.4, >0.6, >0.8, >1, >2, >3, >4, or >5. 0343. In some embodiments, the topoisomerase inhibitor \ CD/ Comonomer is attached to the CDP via a second compound. 0344. In some embodiments, administration of the CDP topoisomerase inhibitor conjugate to a Subject results in D D release of the topoisomerase inhibitor over a period of at least 6 hours. In some embodiments, administration of the CDP 0349 wherein each L is independently a linker, and each D topoisomerase inhibitor conjugate to a Subject results in is independently a topoisomerase inhibitor, a prodrug deriva release of the topoisomerase inhibitor over a period of 2 tive thereof, e.g., a camptothecin or camptothecin derivative, US 2011/O160 159 A1 Jun. 30, 2011 27 or absent; and each comonomer is independently a comono L is one or more of alanine, arginine, histidine, lysine, aspar mer described herein, and n is at least 4, 5, 6, 7, 8, 9, 10, 11, tic acid, glutamic acid, serine, threonine, asparganine, 12, 13, 14, 15, 16, 17, 18, 19 or 20, provided that the polymer glutamine, cysteine, glycine, proline, isoleucine, leucine, comprises at least one topoisomerase inhibitor and in some methionine, phenylalanine, tryptophan, tyrosine and valine. embodiments, at least two topoisomerase inhibitor moieties. In some embodiments, the molecular weight of the comono 0356. In some embodiments, the CDP-topoisomerase mer is from about 2000 to about 5000 Da (e.g., from about inhibitor conjugate is a polymer having attached thereto a 3000 to about 4000 Da (e.g., about 3400 Da). plurality of L-D moieties of the following formula: 0350. In some embodiments, the topoisomerase inhibitor is a topoisomerase inhibitor described herein, for example, the topoisomerase inhibitor is a camptothecin or camptoth ecin derivative described herein. The topoisomerase inhibitor can be attached to the CDP via a functional group such as a hydroxyl group, or where appropriate, an amino group. In Some embodiments, one or more of the topoisomerase inhibi tor moieties in the CDP-topoisomerase inhibitor conjugate can be replaced with another therapeutic agent, e.g., another wherein each L is independently a linker or absent and each D anticancer agent or anti-inflammatory agent. is independently a topoisomerase inhibitor, a prodrug deriva 0351. In some embodiments, the CDP-topoisomerase tive thereof, e.g., a camptothecin or camptothecin derivative, inhibitor conjugate is a polymer having attached thereto a or absent and wherein the group plurality of D moieties of the following formula: -N-1s O -k iii. has a Mw of 3.4 kDa or less and n is at least 4, 5, 6,7,8,9, 10, D D O 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20, provided that the polymer comprises at least one topoisomerase inhibitor and 0352 wherein each L is independently a linker, and each D in Some embodiments, at least two topoisomerase inhibitor is independently a topoisomerase, a prodrug derivative moieties. thereof, e.g., a camptothecin or camptothecin derivative, or absent, provided that the polymer comprises at least one 0357. In some embodiments, less than all of the C(=O) topoisomerase inhibitor and in Some embodiments, at least moieties are attached to L-D moieties, meaning in some two topoisomerase inhibitor moieties; and embodiments, at least one L and/or D is absent. In some 0353 wherein the group embodiments, the loading of the L., D and/or L-D moieties on the CDP-topoisomerase inhibitor conjugate is from about 1 to about 50% (e.g., from about 1 to about 25%, from about 5 to about 20% or from about 5 to about 15%). In some embodi -N-1s ments, each L is independently an amino acid or derivative thereof. In some embodiments, each L is glycine or a deriva has a Mw of 3.4 kDa or less and n is at least 4, 5, 6,7,8,9, 10, tive thereof. 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20. 0358. In some embodiments, one or more of the topoi 0354. In some embodiments, the topoisomerase inhibitor is a topoisomerase inhibitor described herein, for example, somerase inhibitor moieties in the CDP-topoisomerase the topoisomerase is a camptothecin or camptothecin deriva inhibitor conjugate can be replaced with another therapeutic tive described herein. The topoisomerase inhibitor can be agent, e.g., another anticancer agent or anti-inflammatory attached to the CDP via a functional group such as a hydroxyl agent. group, or where appropriate, an amino group. In some 0359. In some embodiments, the CDP-topoisomerase embodiments, one or more of the topoisomerase inhibitor inhibitor conjugate is a polymer having the following for moieties in the CDP-topoisomerase inhibitor conjugate can mula: be replaced with another therapeutic agent, e.g., another anti cancer agent or anti-inflammatory agent. 0355. In some embodiments, less than all of the L moieties are attached to D moieties, meaning in Some embodiments, at least one Disabsent. In some embodiments, the loading of the 4. cyycz, sh-'-r) O D moieties on the CDP-topoisomerase inhibitor conjugate is HN O HN O from about 1 to about 50% (e.g., from about 1 to about 25%, from about 5 to about 20% or from about 5 to about 15%). In s’ os Some embodiments, each L independently comprises an D D amino acid or a derivative thereof. In some embodiments, each Lindependently comprises a plurality of amino acids or derivatives thereof. In some embodiments, each L is indepen 0360. In some embodiments, less than all of the C(=O) dently a dipeptide or derivative thereof. In one embodiment, moieties are attached to US 2011/O160 159 A1 Jun. 30, 2011 28

moiety optionally comprises a linker moiety that forms a covalent linkage between the cyclic structure and the polymer O backbone, preferably having from 1 to 20 atoms in the chain, Such as alkyl chains, including dicarboxylic acid derivatives D lus (such as glutaric acid derivatives, succinic acid derivatives, and the like), and heteroalkyl chains, such as oligoethylene glycol chains. moieties, meaning in some embodiments, 0365 Cyclodextrins are cyclic polysaccharides contain ing naturally occurring D-(+)-glucopyranose units in an O-(1, 4) linkage. The most common cyclodextrins are alpha ((C)- O cyclodextrins, beta (B)-cyclodextrins and gamma (Y)- cyclodextrins which contain, respectively six, seven, or eight Dlunt glucopyranose units. Structurally, the cyclic nature of a cyclo dextrin forms a torus or donut-like shape having an inner is absent, provided that the polymer comprises at least one apolar or hydrophobic cavity, the secondary hydroxyl groups topoisomerase inhibitor and in Some embodiments, at least situated on one side of the cyclodextrin torus and the primary two topoisomerase inhibitor moieties. In some embodiments, hydroxyl groups situated on the other. Thus, using (B)-cyclo the loading of the dextrin as an example, a cyclodextrin is often represented schematically as follows.

OH O D lus O HO OHOo OH moieties on the CDP-topoisomerase inhibitor conjugate is HO Ho' from about 1 to about 50% (e.g., from about 1 to about 25%, O OH from about 5 to about 20% or from about 5 to about 15%). OH O Secondary hydroxyl 0361. In some embodiments, one or more of the topoi O HO ydroxy somerase inhibitor moieties in the CDP-topoisomerase inhibitor conjugate can be replaced with another therapeutic HO OH O V 7 OH agent, e.g., another anticancer agent or anti-inflammatory primary agent. O hydroxyl 0362. In some embodiments, the CDP-topoisomerase HO HO OH inhibitor conjugate will contain an topoisomerase inhibitor O and at least one additional therapeutic agent. For instance, a O OH OHO topoisomerase inhibitor and one more different cancer , an immunosuppressant, an oran anti-inflammatory agent may be grafted on to the polymer via optional linkers. O HO By selecting different linkers for different drugs, the release of each drug may be attenuated to achieve maximal dosage 0366. The side on which the secondary hydroxyl groups and efficacy. are located has a wider diameter than the side on which the 0363 Cyclodextrins primary hydroxyl groups are located. The present invention 0364. In certain embodiments, the cyclodextrin moieties contemplates covalent linkages to cyclodextrin moieties on make up at least about 2%. 5% or 10% by weight, up to 20%, the primary and/or secondary hydroxyl groups. The hydro 30%, 50% or even 80% of the CDP by weight. In certain phobic nature of the cyclodextrin inner cavity allows for embodiments, the topoisomerase inhibitors, or targeting host-guest inclusion complexes of a variety of compounds, ligands make up at least about 1%. 5%, 10% or 15%, 20%, e.g., adamantane. (Comprehensive Supramolecular Chemis 25%, 30% or even 35% of the CDP by weight. Number try, Volume 3, J. L. Atwood et al., eds. Pergamon Press average molecular weight (MO may also vary widely, but (1996); T. Cserhati, Analytical Biochemistry, 225:328-332 generally fall in the range of about 1,000 to about 500,000 (1995); Husain et al., Applied Spectroscopy, 46:652-658 daltons, preferably from about 5000 to about 200,000 daltons (1992); FR 2665 169). Additional methods for modifying and, even more preferably, from about 10,000 to about 100, polymers are disclosed in Suh, J. and Noh, Y. Bioorg. Med. 000. Most preferably, M. varies between about 12,000 and Chem. Lett. 1998, 8, 1327-1330. 65,000 daltons. In certain other embodiments, M. varies 0367. In certain embodiments, the compounds comprise between about 3000 and 150,000 daltons. Within a given cyclodextrin moieties and wherein at least one or a plurality sample of a Subject polymer, a wide range of molecular of the cyclodextrin moieties of the CDP-topoisomerase weights may be present. For example, molecules within the inhibitor conjugate is oxidized. In certain embodiments, the sample may have molecular weights that differ by a factor of cyclodextrin moieties of Palternate with linker moieties in 2, 5, 10, 20, 50, 100, or more, or that differ from the average the polymer chain. molecular weight by a factor of 2, 5, 10, 20, 50, 100, or more. 0368 Comonomers Exemplary cyclodextrin moieties include cyclic structures 0369. In addition to a cyclodextrin moiety, the CDP can consisting essentially of from 7 to 9 saccharide moieties. Such also include a comonomer, for example, a comonomer as cyclodextrin and oxidized cyclodextrin. A cyclodextrin described herein. In some embodiments, a comonomer of the US 2011/O160 159 A1 Jun. 30, 2011 29

CDP-topoisomerase inhibitor conjugate comprises a moiety ticle, e.g., a nanoparticle. The nanoparticle ranges in size selected from the group consisting of an alkylene chain, from 10 to 300 nm in diameter, e.g., 20 to 280, 30 to 250, 40 poly Succinic anhydride, poly-L-glutamic acid, poly(ethyl to 200, 20 to 150, 30 to 100, 20 to 80, 30 to 70, 40 to 60 or 40 eneimine), an oligosaccharide, and an amino acid chain. In to 50 nm diameter. In one embodiment, the particle is 50 to 60 some embodiments, a CDP-topoisomerase inhibitor conju nm, 20 to 60 nm, 30 to 60 nm, 35 to 55 nm, 35 to 50 nm or 35 gate comonomer comprises a polyethylene glycol chain. In to 45 nm in diameter. Some embodiments, a comonomer comprises a moiety selected from: polyglycolic acid and polylactic acid chain. In 0374. In one embodiment, the surface charge of the mol Some embodiments, a comonomer comprises a hydrocarby ecule is neutral, or slightly negative. In some embodiments, lene group wherein one or more methylene groups is option the Zeta potential of the particle surface is from about-80 mV ally replaced by a group Y (provided that none of theY groups to about 50 mV, about -20 mV to about 20 mV, about -20 mV. are adjacent to each other), wherein each Y, independently for to about -10 mV, or about -10 mV to about 0. each occurrence, is selected from, Substituted or unsubsti 0375. In some embodiments, the CDP-topoisomerase tuted aryl, heteroaryl, cycloalkyl, heterocycloalkyl, or inhibitor conjugate is a polymer having the following formula —O , C(=X) (wherein X is NRO or S), —OC(O)—, C:

formula C OH

O OHO

OH O HO HO O OH HO HO O HO O O D OH O HO L --> O -h- iii. OH V . pi HO HO LND O O

O OH OH > O O OH

HO

—C(=O)C), —C(O)NR——S(O) (wherein n is 0, 1, or wherein L and L' independently for each occurrence, is a 2), —OC(O) NR = NR, C(O) NR-, - NR1-C linker, a bond, or —OH and D, independently for each occur (NR) NR—, and —B(OR)—; and R, independently for rence, is a topoisomerase inhibitor Such as camptothecin each occurrence, represents H or a lower alkyl. (“CPT), a camptothecin derivative or absent, and 0370. In some embodiments, a comonomer can be and/or 0376 wherein the group can comprise a linker Such as a linker described herein. 0371 Exemplary CDP-Topoisomerase Inhibitor Conju gates, Particles and Compositions -N1)N 0372. In one embodiment, the CDP-topoisomerase inhibi tor conjugate forms a particle, e.g., a nanoparticle. The par ticle can comprise a CDP-topoisomerase inhibitor conjugate, has a Mw of 3.4 kDa or less and n is at least 4, provided that e.g., a plurality of CDP-topoisomerase inhibitor conjugates, at least one D is CPT or a camptothecin derivative. In some embodiments, at least 2D moieties are CPT and/or a camp e.g., CDP-topoisomerase inhibitor conjugates having the tothecin derivative. same topoisomerase inhibitor or different topoisomerase 0377. In some embodiments, each L', for each occurrence, inhibitors. The compositions described herein comprise a is a cysteine. In some embodiments, the cysteine is attached to CDP-topoisomerase inhibitor conjugate or a plurality of the cyclodextrin via a sulfidebond. In some embodiments, the CDP-topoisomerase inhibitor conjugates. The composition cysteine is attached to the PEG containing portion of the can also comprise a particle or a plurality of particles polymer via an amide bond. described herein. 0378. In some embodiments, the L is a linker (e.g., an 0373) In one embodiment, the CDP-topoisomerase inhibi amino acid such as glycine). In some embodiments, L is tor conjugate containing the inclusion complex forms a par absent. In some embodiments, D-L together form US 2011/O160 159 A1 Jun. 30, 2011 30

and in Some embodiments, at least two

0379. In some embodiments, a plurality of D moieties are absent and at the same position on the polymer, the corre sponding L is —OH. 0380. In some embodiments, less than all of the C(=O) moieties of the cysteine residue in the polymer backbone are attached to moieties. In some embodiments, the loading of the

moieties on the CDP-topoisomerase inhibitor conjugate is from about 1 to about 50% (e.g., from about 1 to about 25%, from about 5 to about 20% or from about 5 to about 15%, e.g., from about 6 to about 10%). In some embodiments, the load ing of

is absent in one or more positions of the polymer backbone, provided that the polymer comprises at least one

NH2

O on the CDP is from about 6% to about 10% by weight of the total polymer. N 0381. In some embodiments, the CDP-topoisomerase inhibitor conjugate of formula C is a polymer having the following formula: US 2011/O160 159 A1 Jun. 30, 2011 31

OH

O OHO OH O - HO Ö OH HO HO O D O HO O L O O OH H

O S N N-1Niii. N S OH HO O O H HO HO O O O pi O OH OH HO O D

O O O OH

HO wherein L, independently for each occurrence, is a linker, a has a Mw of 3.4 kDa or less and n is at least 4, provided that bond,camptothecin or—C Hand (“CPT), P. independently a camptothecin for derivative each occurrence, or absent, is at least one D is CPT or a camptothecin derivative. In some and embodiments, at least 2D moieties are CPT and/or a camp wherein the group tothecin derivative.

0382. In some embodiments, the CDP-camptothecin con jugate of formula C is a polymer of the following formula:

US 2011/O160 159 A1 Jun. 30, 2011 32

0383 wherein m and n are as defined above, and wherein (0389. Described herein are CDP-topoisomerase inhibitor less than all of the C(=O) sites of the cysteine of the polymer conjugates comprising a CDP covalently attached to a topoi backbone are occupied as indicated above with the CPT-Gly, Somerase inhibitor through attachments that are cleaved but instead are free acids, meaning, the theoretical loading of under biological conditions to release the topoisomerase the polymer is less than 100%. inhibitor. In certain embodiments, a CDP-topoisomerase 0384. In some embodiments, the CDP-camptothecin con inhibitor conjugate comprises a topoisomerase inhibitor jugate is as provided in FIG. 4, and shown below, which is covalently attached to a polymer, preferably a biocompatible referred to herein as “CRLX101. polymer, through a tether, e.g., a linker, wherein the tether

OH

In the above structure: comprises a selectivity-determining moiety and a self-cycliz n=about 77 or the molecular weight of the PEG moiety is ing moiety which are covalently attached to one another in the from about 3060 to about 3740 (e.g., about 3400) Da: tether, e.g., between the polymer and the topoisomerase m is from about 10 to about 18 (e.g., about 14); inhibitor. the molecular weight of the polymer backbone (i.e., the poly 0390. In some embodiments, such topoisomerase inhibi mer minus the CPT-gly, which results in the cysteine moieties tors are covalently attached to CDPs through functional having a free —C(O)OH) is from about 48 to about 85 kDa, groups comprising one or more heteroatoms, for example, the polydispersity of the polymer backbone is less than about hydroxy, thiol, carboxy, amino, and amide groups. Such 2.2; and groups may be covalently attached to the Subject polymers the loading of the CPT onto the polymer backbone is from through linker groups as described herein, for example, bio about 6 to about 13% by weight, wherein 13% is theoretical cleavable linker groups, and/or through tethers, such as a maximum, meaning, in Some instances, one or more of the tether comprising a selectivity-determining moiety and a self cysteine residues has a free —C(O)OH (i.e., it lacks the cyclizing moiety which are covalently attached to one CPT-gly). another. 0385. In some embodiments, the polydispersity of the 0391. In certain embodiments, the CDP-topoisomerase PEG component in the above structure is less than about 1.1. inhibitor conjugate comprises a topoisomerase inhibitor 0386. In some embodiments, a CDP-camptothecin conju covalently attached to the CDP through a tether, wherein the gate described herein has a terminal and/or a terminal tether comprises a self-cyclizing moiety. In some embodi carboxylic acid. ments, the tether further comprises a selectivity-determining moiety. Thus, one aspect of the invention relates to a polymer Linkers/Tethers conjugate comprising a topoisomerase inhibitor covalently (0387. The CDPs described herein can include on or more attached to a polymer, preferably a biocompatible polymer, linkers. In some embodiments, a linker can link a topoi through a tether, wherein the tether comprises a selectivity somerase inhibitor to a CDP. In some embodiments, a linker determining moiety and a self-cyclizing moiety which are can link camptothecin or a camptothecin derivative to a CDP. covalently attached to one another. In some embodiments, for example, when referring to a linker 0392. In some embodiments, the selectivity-determining that links a topoisomerase inhibitor to the CDP the linker can moiety is bonded to the self-cyclizing moiety between the be referred to as a tether. self-cyclizing moiety and the CDP. 0388. In certain embodiments, a plurality of the linker 0393. In certain embodiments, the selectivity-determining moieties are attached to a topoisomerase inhibitor or prodrug moiety is a moiety that promotes selectivity in the cleavage of thereof and are cleaved under biological conditions. the bond between the selectivity-determining moiety and the US 2011/O160 159 A1 Jun. 30, 2011 self-cyclizing moiety. Such a moiety may, for example, pro prising one or more methylene groups, wherein one or more mote enzymatic cleavage between the selectivity-determin methylene groups is optionally replaced by a group Y (pro ing moiety and the self-cyclizing moiety. Alternatively, Such vided that none of the Y groups are adjacent to each other), a moiety may promote cleavage between the selectivity-de wherein each Y, independently for each occurrence, is termining moiety and the self-cyclizing moiety under acidic selected from, substituted or unsubstituted aryl, heteroaryl, conditions or basic conditions. cycloalkyl, heterocycloalkyl, or —O—, C(=X) (wherein X 0394. In certain embodiments, the invention contemplates is NR', O or S), OC(O) C(=O)C), NR , any combination of the foregoing. Those skilled in the art will recognize that, for example, any topoisomerase inhibitor of - NRCO - C(O)NR' , S(O), (wherein n is 0, 1, the invention in combination with any linker (e.g., self-cy or 2), OC(O) NR, NR C(O) NR , clizing moiety, any selectivity-determining moiety, and/or NR C(NR) NRO , and B(OR) ; and R, any topoisomerase inhibitor) are within the scope of the independently for each occurrence, represents H or a lower invention. alkyl. In certain embodiments, J may be substituted or unsub 0395. In certain embodiments, the selectivity-determining stituted lower alkylene, such as ethylene. For example, the moiety is selected such that the bond is cleaved under acidic selectivity-determining moiety may be conditions. 0396. In certain embodiments, where the selectivity-deter mining moiety is selected Such that the bond is cleaved under H basic conditions, the selectivity-determining moiety is an aminoalkylcarbonyloxyalkyl moiety. In certain embodi ~y ments, the selectivity-determining moiety has a structure 0402. In certain embodiments, the selectivity-determining O moiety is represented by Formula B:

O var- y (B) 0397. In certain embodiments where the selectivity-deter W 1. J Ns 1 S N 1 Q y mining moiety is selected Such that the bond is cleaved enzy matically, it may be selected Such that a particular or class of cleaves the bond. In certain preferred Such embodiments, the selectivity-determining moiety may be wherein selected such that the bond is cleaved by a cathepsin, prefer ably cathepsin B. W is either a direct bond or selected from lower alkyl, NR'', 0398. In certain embodiments the selectivity-determining S, O: moiety comprises a peptide, preferably a dipeptide, tripep S is sulfur, tide, or tetrapeptide. In certain such embodiments, the peptide J, independently and for each occurrence, is hydrocarbyl or is a dipeptide is selected from KF and FK. In certain embodi polyethylene glycol; ments, the peptide is a tripeptide is selected from GFA, GLA, Q is O or NR", wherein R' is hydrogen or alkyl; and AVA, GVA, GIA, GVL, GVF, and AVF. In certain embodi ments, the peptide is a tetrapeptide selected from GFYA and R" is selected from hydrogen and alkyl. GFLG, preferably GFLG. 0403. In certain such embodiments, J may be substituted 0399. In certain such embodiments, a peptide, such as or unsubstituted lower alkyl, such as methylene. In certain GFLG, is selected such that the bond between the selectivity Such embodiments, J may be an aryl ring. In certain embodi determining moiety and the self-cyclizing moiety is cleaved ments, the aryl ring is a benzo ring. In certain embodiments W by a cathepsin, preferably cathepsin B. and S are in a 1.2-relationship on the aryl ring. In certain 0400. In certain embodiments, the selectivity-determining embodiments, the aryl ring may be optionally Substituted moiety is represented by Formula A: with alkyl, alkenyl, alkoxy, aralkyl, aryl, heteroaryl, halogen, CN, azido, NR'R, COOR, C(O) NR'R', C(O) R, NR C(O) R, NRSOR', SR, (A) S(O)R', SOR', SONR'R'', -(C(R).), OR, —(C(R)), NR'R', and —(C(R)). SOR: wherein R is, independently for each occurrence, H or lower alkyl, and in is, independently for each occurrence, an integer from 0 to 2. ---- 04.04. In certain embodiments, the aryl ring is optionally Substituted with alkyl, alkenyl, alkoxy, aralkyl, aryl, het wherein eroaryl, halogen, —CN, azido, NR'R''. —COOR, Sa sulfur atom that is part of a disulfide bond; C(O) NR'R', C(O) R', NR C(O) R, J is optionally substituted hydrocarbyl; and NRSOR', SR, S(O)R, -SOR', -SONR'R', Q is O or NR", wherein R' is hydrogen or alkyl. —(C(R)), OR, —(C(R)), NR'R'', and —(C(R).) 04.01. In certain embodiments, J may be polyethylene gly SOR: wherein R is, independently for each occurrence, col, polyethylene, polyester, alkenyl, or alkyl. In certain H or lower alkyl, and n is, independently for each occurrence, embodiments, J may represent a hydrocarbylene group com an integer from 0 to 2. US 2011/O160 159 A1 Jun. 30, 2011 34

0405. In certain embodiments, J, independently and for each occurrence, is polyethylene glycol, polyethylene, poly ester, alkenyl, or alkyl. 0406. In certain embodiments, independently and for each S occurrence, the linker comprises a hydrocarbylene group O s1 N-X. comprising one or more methylene groups, wherein one or more methylene groups is optionally replaced by a group Y (provided that none of the Y groups are adjacent to each other), wherein each Y, independently for each occurrence, is selected from, substituted or unsubstituted aryl, heteroaryl, 0411. In certain embodiments, the self-cyclizing moiety is cycloalkyl, heterocycloalkyl, or —O—, C(=X) (wherein X selected such that upon cleavage of the bond between the is NR', O or S), —OC(O) , —C(=O)C), NRCO , selectivity-determining moiety and the self-cyclizing moiety, —C(O)NR' , S(O), (wherein n is 0, 1, or 2), —OC cyclization occurs thereby releasing the therapeutic agent. (O) NR, NRO C(O) NRO , NRO C Such a cleavage-cyclization-release cascade may occur (NR) NR' , and B(OR) ; and R', indepen sequentially in discrete steps or Substantially simultaneously. dently for each occurrence, represents H or a lower alkyl. Thus, in certain embodiments, there may be a temporal and/or 0407. In certain embodiments, J, independently and for spatial difference between the cleavage and the self-cycliza tion. The rate of the self-cyclization cascade may depend on each occurrence, is Substituted or unsubstituted lower alky pH, e.g., a basic pH may increase the rate of self-cyclization lene. In certain embodiments, J., independently and for each after cleavage. Self-cyclization may have a half-life after occurrence, is substituted or unsubstituted ethylene. introduction in vivo of 24 hours, 18 hours, 14 hours, 10 hours, 0408. In certain embodiments, the selectivity-determining 6 hours, 3 hours, 2 hours, 1 hour, 30 minutes, 10 minutes, 5 moiety is selected from minutes, or 1 minute. 0412. In certain such embodiments, the self-cyclizing moiety may be selected Such that, upon cyclization, a five- or six-membered ring is formed, preferably a five-membered ring. In certain such embodiments, the five- or six-membered ring comprises at least one heteroatom selected from oxygen, as-s-s-X and , or sulfur, preferably at least two, wherein the het O eroatoms may be the same or different. In certain Such H embodiments, the heterocyclic ring contains at least one nitrogen, preferably two. In certain such embodiments, the self-cyclizing moiety cyclizes to form an imidazolidone. A-s-s-X.O 0413. In certain embodiments, the self-cyclizing moiety has a structure The selectivity-determining moiety may include groups with bonds that are cleavable under certain conditions, such as R2 disulfide groups. In certain embodiments, the selectivity-de V. X s termining moiety comprises a disulfide-containing moiety, U for example, comprising aryland/or alkyl group(s) bonded to a disulfide group. In certain embodiments, the selectivity R3 O determining moiety has a structure wherein U is selected from NR' and S; O R20 X is selected from O, NR, and S, preferably O or S; V is selected from O, S and NR", preferably O or NR; - - - R° and Rare independently selected from hydrogen, alkyl, O 1Ns 1 s’, and alkoxy; or R and R together with the atoms to which they are attached form a ring; and R", R, and Rare independently selected from hydrogen and wherein alkyl. 0414. In certain embodiments, U is NR' and/or V is NR, Ar is a Substituted or unsubstituted benzo ring; and R' and Rare independently selected from methyl, ethyl, J is optionally substituted hydrocarbyl; and propyl, and isopropyl. In certain embodiments, both R' and Rare methyl. On certain embodiments, both R and Rare Q is O or NR', hydrogen. In certain embodiments R and R are indepen dently alkyl, preferably lower alkyl. In certain embodiments, 04.09 wherein R' is hydrogen or alkyl. R° and R together are —(CH), wherein n is 3 or 4. 0410. In certain embodiments, Aris unsubstituted. In cer thereby forming a cyclopentyl or cyclohexyl ring. In certain tain embodiments, Aris a 12-benzo ring. For example, Suit embodiments, the nature of R and R may affect the rate of able moieties within Formula B include: cyclization of the self-cyclizing moiety. In certain Such US 2011/O160 159 A1 Jun. 30, 2011 embodiments, it would be expected that the rate of cyclization a leaving group linker, such as CO or methoxymethyl, that would be greater when R and R together with the carbon spontaneously dissociates from the remainder of the mol atoms to which they are attached form a ring than the rate ecule after cleavage occurs. when R and Rare independently selected from hydrogen, 0419. In some embodiments, a linker may be and/or com alkyl, and alkoxy. In certain embodiments, U is bonded to the prise an alkylene chain, a polyethylene glycol (PEG) chain, self-cyclizing moiety. poly Succinic anhydride, poly-L-glutamic acid, poly(ethyl 0415. In certain embodiments, the self-cyclizing moiety is eneimine), an oligosaccharide, an amino acid (e.g., glycine or selected from cysteine), an amino acid chain, or any other Suitable linkage. In certain embodiments, the linker group itself can be stable under physiological conditions, such as an alkylene chain, or it can be cleavable under physiological conditions, such as by an enzyme (e.g., the linkage contains a peptide sequence that is a Substrate for a peptidase), or by hydrolysis (e.g., the linkage contains a hydrolyzable group, Such as an ester or thioester). The linker groups can be biologically inactive, Such as a PEG, polyglycolic acid, or polylactic acid chain, or can be biologically active, Such as an oligo- or polypeptide that, when cleaved from the moieties, binds a receptor, deac tivates an enzyme, etc. Various oligomeric linker groups that are biologically compatible and/or bioerodible are known in the art, and the selection of the linkage may influence the ultimate properties of the material, such as whether it is durable when implanted, whether it gradually deforms or shrinks after implantation, or whether it gradually degrades and is absorbed by the body. The linkergroup may be attached to the moieties by any suitable bond or functional group, including carbon-carbon bonds, esters, ethers, amides, , carbonates, carbamates, Sulfonamides, etc. 0420. In certain embodiments, the linker group(s) of the 0416) In certain embodiments, the selectivity-determining present invention represent a hydrocarbylene group wherein moiety may connect to the self-cyclizing moiety through one or more methylene groups is optionally replaced by a group Y (provided that none of the Y groups are adjacent to carbonyl-heteroatom bonds, e.g., amide, carbamate, carbon each other), wherein each Y, independently for each occur ate, ester, thioester, and bonds. rence, is selected from, substituted or unsubstituted aryl, het 0417. In certain embodiments, a topoisomerase inhibitor eroaryl, cycloalkyl, heterocycloalkyl, or —O—, C(=X) is covalently attached to a polymer through a tether, wherein the tether comprises a selectivity-determining moiety and a (wherein X is NR, O or S), OC(O)—, C(=O)C), self-cyclizing moiety which are covalently attached to one NRCO , —C(O)NR— —S(O), (wherein n is 0, 1, another. In certain embodiments, the self-cyclizing moiety is or 2), —OC(O) NR, NR-C(O) NR-, - NR-C selected such that after cleavage of the bond between the (NR) NR—, and —B(OR)—; and R, independently for selectivity-determining moiety and the self-cyclizing moiety, each occurrence, represents H or a lower alkyl. cyclization of the self-cyclizing moiety occurs, thereby 0421. In certain embodiments, the linker group represents releasing the therapeutic agent. As an illustration, ABC may a derivatized or non-derivatized amino acid (e.g., glycine or be a selectivity-determining moiety, and DEFGH maybe be a cysteine). In certain embodiments, linker groups with one or self-cyclizing moiety, and ABC may be selected Such that more terminal carboxyl groups may be conjugated to the enzyme Y cleaves between C and D. Once cleavage of the polymer. In certain embodiments, one or more of these ter bond between C and D progresses to a certain point, D will minal carboxyl groups may be capped by covalently attaching cyclize onto H, thereby releasing topoisomerase inhibitor X, them to a therapeutic agent, a targeting moiety, or a cyclo dextrin moiety via an (thio)ester or amide bond. In still other or a prodrug thereof. embodiments, linker groups with one or more terminal hydroxyl, thiol, oramino groups may be incorporated into the polymer. In preferred embodiments, one or more of these terminal hydroxyl groups may be capped by covalently A- B D NE1 FNo1 H Nx - - attaching them to a therapeutic agent, a targeting moiety, or a cyclodextrin moiety viaan (thio)ester, amide, carbonate, car bamate, thiocarbonate, or thiocarbamate bond. In certain embodiments, these (thio)ester, amide, (thio)carbonate or (thio)carbamates bonds may be biohydrolyzable, i.e., capable of being hydrolyzed under biological conditions. 0422. In certain embodiments, a linker group represents a hydrocarbylene group wherein one or more methylene groups is optionally replaced by a group Y (provided that 0418. In certain embodiments, topoisomerase inhibitor X none of the Ygroups are adjacent to each other), wherein each may further comprise additional intervening components, Y. independently for each occurrence, is selected from, sub including, but not limited to another self-cyclizing moiety or stituted or unsubstituted aryl, heteroaryl, cycloalkyl, hetero US 2011/O160 159 A1 Jun. 30, 2011 36 cycloalkyl, or —O C(=X) (wherein X is NR, O or S), 0433. In certain embodiments, the linker group comprises –OC(O) , C(=O)C), NRCO -, - C(O)NR-, an amino acid or peptide, or derivative thereof (e.g., a glycine —S(O), (wherein n is 0, 1, or 2), —OC(O)—NR, or cysteine). NR—C(O) NR-, NR—C(NR) NR—, and 0434. In certain embodiments as disclosed herein, the —B(OR)—, and R, independently for each occurrence, linker is connected to the topoisomerase inhibitor through a represents H or a lower alkyl. hydroxyl group. In certain embodiments as disclosed herein, 0423 In certain embodiments, a linker group, e.g., the linker is connected to the topoisomerase inhibitor through between a topoisomerase inhibitor and the CDP. comprises a an amino group. self-cyclizing moiety. In certain embodiments, a linker group, 0435. In certain embodiments, the linker group that con e.g., between a topoisomerase inhibitor and the CDP com nects to the topoisomerase inhibitor may comprise a self prises a selectivity-determining moiety. cyclizing moiety, or a selectivity-determining moiety, or 0424. In certain embodiments as disclosed herein, a linker both. In certain embodiments, the selectivity-determining group, e.g., between a topoisomerase inhibitor and the CDP. moiety is a moiety that promotes selectivity in the cleavage of comprises a self-cyclizing moiety and a selectivity-determin the bond between the selectivity-determining moiety and the ing moiety. self-cyclizing moiety. Such a moiety may, for example, pro 0425. In certain embodiments as disclosed herein, the mote enzymatic cleavage between the selectivity-determin topoisomerase inhibitor or targeting ligand is covalently ing moiety and the self-cyclizing moiety. Alternatively, Such bonded to the linker group via a biohydrolyzable bond (e.g., a moiety may promote cleavage between the selectivity-de an ester, amide, carbonate, carbamate, or a phosphate). termining moiety and the self-cyclizing moiety under acidic 0426 In certain embodiments as disclosed herein, the conditions or basic conditions. CDP comprises cyclodextrin moieties that alternate with 0436. In certain embodiments, any of the linker groups linker moieties in the polymer chain. may comprise a self-cyclizing moiety or a selectivity-deter 0427. In certain embodiments, the linker moieties are mining moiety, or both. In certain embodiments, the selectiv attached to topoisomerase inhibitors or prodrugs thereof that ity-determining moiety may be bonded to the self-cyclizing are cleaved under biological conditions. moiety between the self-cyclizing moiety and the polymer. 0437. In certain embodiments, any of the linker groups 0428. In certain embodiments, at least one linker that con may independently be or include an alkyl chain, a polyethyl nects the topoisomerase inhibitor or prodrug thereof to the ene glycol (PEG) chain, poly Succinic anhydride, poly-L- polymer comprises a group represented by the formula glutamic acid, poly(ethyleneimine), an oligosaccharide, an amino acid chain, or any other Suitable linkage. In certain O embodiments, the linker group itself can be stable under | R41 physiological conditions, such as an alkyl chain, or it can be – E.-K-N cleavable under physiological conditions, such as by an enzyme (e.g., the linkage contains a peptide sequence that is X s a substrate for a peptidase), or by hydrolysis (e.g., the linkage contains a hydrolyzable group. Such as an ester or thioester). wherein The linker groups can be biologically inactive, such as a PEG, P is phosphorus: polyglycolic acid, or polylactic acid chain, or can be biologi O is oxygen; cally active, such as an oligo- or polypeptide that, when cleaved from the moieties, binds a receptor, deactivates an E represents oxygen or NR': enzyme, etc. Various oligomeric linker groups that are bio K represents hydrocarbyl, logically compatible and/or bioerodible are known in the art, X is selected from OR or NR'R'': and and the selection of the linkage may influence the ultimate R", R', R. R', and R dependently represent hydrogen properties of the material, such as whether it is durable when or optionally substituted alkyl. implanted, whether it gradually deforms or shrinks after 0429. In certain embodiments, E is NRandR is hydro implantation, or whether it gradually degrades and is gen. absorbed by the body. The linker group may be attached to the 0430. In certain embodiments, K is lower alkylene (e.g., moieties by any suitable bond or functional group, including ethylene). carbon-carbon bonds, esters, ethers, amides, amines, carbon ates, carbamates, Sulfonamides, etc. 0431. In certain embodiments, at least one linker com 0438. In certain embodiments, any of the linker groups prises a group selected from may independently be an alkyl group wherein one or more methylene groups is optionally replaced by a group Y (pro O H vided that none of the Y groups are adjacent to each other), N wherein each Y, independently for each occurrence, is selected from aryl, heteroaryl, carbocyclyl, heterocyclyl, or and OH -O-, C(=X) (wherein X is NR', O or S), —OC(O) , O - C(=O)C) , – NR'-, - NRCO -, -C(O)NR'-, H —S(O), (wherein n is 0, 1, or 2), —OC(O) NR' , NR' C(O) NR' , NR' C(NR') NR' , and |-r-y —B(OR") ; and R', independently for each occurrence, is OCH2CH3 H or lower alkyl. 0439. In certain embodiments, the present invention con 0432. In certain embodiments, X is OR'. templates a CDP, wherein a plurality of topoisomerase inhibi US 2011/O160 159 A1 Jun. 30, 2011 37 tors are covalently attached to the polymer through attach 0449 CDP-topoisomerase inhibitor conjugates, particles ments that are cleaved under biological conditions to release and compositions of the present invention may be useful to the therapeutic agents as discussed above, wherein adminis improve solubility and/or stability of the topoisomerase tration of the polymer to a subject results in release of the inhibitor, reduce drug-drug interactions, reduce interactions therapeutic agent over a period of at least 2, 3, 5, 6, 8, 10, 15, with blood elements including plasma proteins, reduce or 20, 24, 36, 48 or even 72 hours. eliminate immunogenicity, protect the topoisomerase inhibi 0440. In some embodiments, the conjugation of the topoi tor from metabolism, modulate drug-release kinetics, somerase inhibitor to the CDP improves the aqueous solubil improve circulation time, improve topoisomerase inhibitor ity of the topoisomerase inhibitor and hence the bioavailabil half-life (e.g., in the serum, or in selected tissues, such as ity. Accordingly, in one embodiment of the invention, the tumors), attenuate toxicity, improve efficacy, normalize topoisomerase inhibitor has a log P-0.4, >0.6, >0.8, >1, >2. topoisomerase inhibitor metabolism across subjects of differ >3, >4, or even >5. ent , ethnicities, and/or races, and/or provide for tar 0441 The CDP-topoisomerase inhibitor conjugate of the geted delivery into specific cells or tissues. present invention preferably has a molecular weight in the 0450. In other embodiments, the CDP-topoisomerase range of 10,000 to 500,000; 30,000 to 200,000; or even inhibitor conjugate, particle or composition may be a flexible 70,000 to 150,000 amu. or flowable material. When the CDP used is itself flowable, 0442. In certain embodiments, the present invention con the CDP composition of the invention, even when viscous, templates attenuating the rate of release of the topoisomerase need not include a biocompatible solvent to be flowable, inhibitor by introducing various tether and/or linking groups although trace or residual amounts of biocompatible solvents between the therapeutic agent and the polymer. Thus, in cer may still be present. tain embodiments, the CDP-topoisomerase inhibitor conju 0451 While it is possible that the biodegradable polymer gates of the present invention are compositions for controlled or the biologically active agent may be dissolved in a small delivery of the topoisomerase inhibitor. quantity of a solvent that is non-toxic to more efficiently 0443 CDP-Topoisomerase Inhibitor Conjugate Charac produce an amorphous, monolithic distribution or a fine dis teristics persion of the biologically active agent in the flexible or 0444. In some embodiments, the CDP and/or CDP-topoi flowable composition, it is an advantage of the invention that, Somerase inhibitor conjugate, particle or composition as in a preferred embodiment, no solvent is needed to form a described herein have polydispersities less than about 3, or flowable composition. Moreover, the use of solvents is pref even less than about 2. erably avoided because, once a polymer composition contain 0445 One embodiment of the present invention provides ing solvent is placed totally or partially within the body, the an improved delivery of certain topoisomerase inhibitor by Solvent dissipates or diffuses away from the polymer and covalently attaching one or more topoisomerase inhibitors to must be processed and eliminated by the body, placing an a CDP. Such conjugation can improve the aqueous solubility extra burden on the body's clearance ability at a time when the and hence the bioavailability of the topoisomerase inhibitor. illness (and/or other treatments for the illness) may have 0446. The CDP-topoisomerase inhibitor conjugates, par already deleteriously affected it. ticles and compositions described herein preferably have 0452. However, when a solvent is used to facilitate mixing molecular weights in the range of 10,000 to 500,000; 30,000 or to maintain the flowability of the CDP-topoisomerase to 200,000; or even 70,000 to 150,000 amu. In certain inhibitor conjugate, particle or composition, it should be non embodiments as disclosed herein, the compound has a num toxic, otherwise biocompatible, and should be used in rela ber average (M) molecular weight between 1,000 to 500,000 tively small amounts. Solvents that are toxic should not be amu, or between 5,000 to 200,000 amu, or between 10,000 to used in any material to be placed even partially within a living 100,000 amu. One method to determine molecular weight is body. Such a solvent also must not cause Substantial tissue by gel permeation chromatography (“GPC), e.g., mixedbed irritation or necrosis at the site of administration. columns. CHCl solvent, light scattering detector, and off 0453 Examples of suitable biocompatible solvents, when line doidc. Other methods are known in the art. used, include N-methyl-2-pyrrolidone, 2-pyrrolidone, etha 0447. In certain embodiments as disclosed herein, the nol, propylene glycol, acetone, methyl acetate, ethyl acetate, CDP-topoisomerase inhibitor conjugate, particle or compo methyl ethylketone, dimethylformamide, dimethylsulfoxide, sition is biodegradable or bioerodable. tetrahydrofuran, caprolactam, oleic acid, or 1-dodecylazacyl 0448. In certain embodiments as disclosed herein, the coheptanone. Preferred solvents include N-methylpyrroli topoisomerase inhibitor, e.g., the camptothecin, camptoth done, 2-pyrrolidone, dimethylsulfoxide, and acetone because ecin derivative, or prodrug thereof makes up at least 3% (e.g., of their solvating ability and their biocompatibility. at least about 5%) by weight of the polymer. In certain 0454. In certain embodiments, the CDP-topoisomerase embodiments, the topoisomerase inhibitor, e.g., the camp inhibitor conjugates, particles and compositions are soluble tothecin, camptothecin derivative or prodrug thereof makes in one or more common organic solvents for ease of fabrica up at least 20% by weight of the compound. In certain tion and processing. Common organic solvents include Such embodiments, the topoisomerase inhibitor, e.g., the camp Solvents as chloroform, dichloromethane, dichloroethane, tothecin, camptothecin derivative or prodrug thereof makes 2-butanone, butyl acetate, ethyl butyrate, acetone, ethyl up at least 5%, 10%, 15%, or at least 20% by weight of the acetate, dimethylacetamide, N-methylpyrrolidone, dimethyl compound. formamide, and dimethylsulfoxide. US 2011/O160 159 A1 Jun. 30, 2011

0455. In certain embodiments, the CDP-topoisomerase 0461 CDP-topoisomerase inhibitor conjugate, particle or inhibitor conjugates, particles and compositions described composition releases incorporated material at a rate from herein, upon contact with body fluids, undergo gradual deg about 2 or less to about 1000 or more times faster than another radation. The life of a biodegradable polymer in vivo depends polymeric system. upon, among other things, its molecular weight, crystallinity, 0462 Alternatively, a comparison may reveal a rate differ biostability, and the degree of crosslinking. In general, the ence of about 3, 5, 7, 10, 25, 50, 100, 250, 500 or 750 times. greater the molecular weight, the higher the degree of crys Even higher rate differences are contemplated by the present tallinity, and the greater the biostability, the slower biodegra invention and release rate protocols. dation will be. 0463. In certain embodiments, when formulated in a cer 0456. If a subject composition is formulated with a topoi tain manner, the release rate for CDP-topoisomerase inhibitor somerase inhibitor or other material, release of the topoi conjugates, particles and compositions of the present inven somerase inhibitor or other material for a sustained or tion may present as mono- or bi-phasic. extended period as compared to the release from an isotonic 0464 Release of any material incorporated into the poly saline solution generally results. Such release profile may mer matrix, which is often provided as a microsphere, may be result in prolonged delivery (over, say 1 to about 2,000 hours, characterized in certain instances by an initial increased or alternatively about 2 to about 800 hours) of effective release rate, which may release from about 5 to about 50% or amounts (e.g., about 0.0001 mg/kg/hour to about 10 mg/kg/ more of any incorporated material, or alternatively about 10, hour, e.g., 0.001 mg/kg/hour, 0.01 mg/kg/hour, 0.1 mg/kg/ 15, 20, 25, 30 or 40%, followed by a release rate of lesser hour, 1.0 mg/kg/hour) of the topoisomerase inhibitor or any magnitude. other material associated with the polymer. 0465. The release rate of any incorporated material may 0457. A variety of factors may affect the desired rate of also be characterized by the amount of such material released hydrolysis of CDP-topoisomerase inhibitor conjugates, par per day per mg of polymer matrix. For example, in certain ticles and compositions, the desired softness and flexibility of embodiments, the release rate may vary from about 1 ng or the resulting solid matrix, rate and extent of bioactive material less of any incorporated material per day per mg of polymeric release. Some of such factors include the selection/identity of system to about 500 or more ng/day/mg. Alternatively, the the various subunits, the enantiomeric or diastereomeric release rate may be about 0.05, 0.5, 5, 10, 25, 50, 75, 100, 125, purity of the monomeric subunits, homogeneity of subunits 150, 175, 200, 250, 300, 350, 400, 450, or 500 ng/day/mg. In found in the polymer, and the length of the polymer. For still other embodiments, the release rate of any incorporated instance, the present invention contemplates heteropolymers material may be 10,000 ng/day/mg, or even higher. In certain with varying linkages, and/or the inclusion of other mono instances, materials incorporated and characterized by Such meric elements in the polymer, in order to control, for release rate protocols may include therapeutic agents, fillers, example, the rate of biodegradation of the matrix. and other Substances. 0458. To illustrate further, a wide range of degradation 0466. In another aspect, the rate of release of any material rates may be obtained by adjusting the hydrophobicities of from any CDP-topoisomerase inhibitor conjugate, particle or the backbones or side chains of the polymers while still main composition of the present invention may be presented as the taining sufficient biodegradability for the use intended for any half-life of such material in the matrix. Such polymer. Such a result may be achieved by varying the 0467. In addition to the embodiment involving protocols various functional groups of the polymer. For example, the for in vitro determination of release rates, in vivo protocols, combination of a hydrophobic backbone and a hydrophilic whereby in certain instances release rates for polymeric sys linkage produces heterogeneous degradation because cleav tems may be determined in vivo, are also contemplated by the age is encouraged whereas water penetration is resisted. present invention. Other assays useful for determining the 0459. One protocol generally accepted in the field that release of any material from the polymers of the present may be used to determine the release rate of a therapeutic system are known in the art. agent Such as a topoisomerase inhibitor or other material 0468 Physical Structures of the CDP-Topoisomerase loaded in the CDP-topoisomerase inhibitor conjugates, par Inhibitor Conjugates, Particles and Compositions ticles or compositions of the present invention involves deg 0469. The CDP-topoisomerase inhibitor conjugates, par radation of any such matrix in a 0.1 MPBS solution (pH 7.4) ticles and compositions may beformed in a variety of shapes. at 37° C., an assay known in the art. For purposes of the For example, in certain embodiments, CDP-topoisomerase present invention, the term “PBS protocol is used herein to inhibitor conjugates may be presented in the form of micro refer to such protocol. particles or nanoparticles. Microspheres typically comprise a 0460. In certain instances, the release rates of different biodegradable polymer matrix incorporating a drug. Micro CDP-topoisomerase inhibitor conjugates, particles and com spheres can beformed by a wide variety oftechniques known positions of the present invention may be compared by Sub to those of skill in the art. Examples of microsphere forming jecting them to Such a protocol. In certain instances, it may be techniques include, but are not limited to, (a) phase separation necessary to process polymeric systems in the same fashion to by emulsification and Subsequent organic solvent evaporation allow direct and relatively accurate comparisons of different (including complex emulsion methods such as oil in water systems to be made. For example, the present invention emulsions, water in oil emulsions and water-oil-water emul teaches several different methods of formulating the CDP sions); (b) coacervation-phase separation; (c) melt disper topoisomerase inhibitor conjugates, particles and composi sion; (d) interfacial deposition; (e) in situ polymerization, (f) tions. Such comparisons may indicate that any one spray drying and spray congealing; (g) air Suspension coat US 2011/O160 159 A1 Jun. 30, 2011 39 ing; and (h) pan and spray coating. These methods, as well as 0475. The CDPs described herein can be made using a properties and characteristics of microspheres are disclosed variety of methods including those described herein. In some in, for example, U.S. Pat. No. 4,438,253: U.S. Pat. No. 4,652, embodiments, a CDP can be made by: providing cyclodextrin 441; U.S. Pat. No. 5,100,669; U.S. Pat. No. 5,330,768: U.S. moiety precursors; providing comonomer precursors which Pat. No. 4,526,938; U.S. Pat. No. 5,889, 110; U.S. Pat. No. do not contain cyclodextrin moieties (comonomer precur 6,034,175; and European Patent 0258780, the entire disclo sors); and copolymerizing the said cyclodextrin moiety pre sures of which are incorporated by reference herein in their cursors and comonomer precursors to thereby make a CDP entireties. wherein CDP comprises at least four cyclodextrin moieties 0470 To prepare microspheres, several methods can be and at least four comonomers. employed depending upon the desired application of the 0476. In some embodiments, the at least four cyclodextrin delivery vehicles. Suitable methods include, but are not lim moieties and at least four comonomers alternate in the water ited to, spray drying, freeze drying, air drying, vacuum dry soluble linear polymer. In some embodiments, the method ing, fluidized-bed drying, milling, co-precipitation and criti includes providing cyclodextrin moiety precursors modified cal fluid extraction. In the case of spray drying, freeze drying, to bear one reactive site at each of exactly two positions, and air drying, vacuum drying, fluidized-bed drying and critical reacting the cyclodextrin moiety precursors with comonomer fluid extraction; the components (stabilizing polyol, bioactive precursors having exactly two reactive moieties capable of material, buffers, etc.) are first dissolved or suspended in forming a covalent bond with the reactive sites under poly aqueous conditions. In the case of milling, the components merization conditions that promote reaction of the reactive are mixed in the dried form and milled by any method known sites with the reactive moieties to form covalent bonds in the art. In the case of co-precipitation, the components are between the comonomers and the cyclodextrin moieties, mixed in organic conditions and processed as described whereby a CDP comprising alternating units of a cyclodextrin below. Spray drying can be used to load the stabilizing polyol moiety and a comonomer is produced. with the bioactive material. The components are mixed under 0477. In some embodiments, the cyclodextrin momomers aqueous conditions and dried using precision nozzles to pro comprise linkers to which the topoisomerase inhibitor may be duce extremely uniform droplets in a drying chamber. Suit further linked able spray drying machines include, but are not limited to, 0478 In some embodiments, the comonomer precursor is Buchi, NIRO, APV and Lab-plant spray driers used according a compound containing at least two functional groups to the manufacturer's instructions. through which reaction and thus linkage of the cyclodextrin 0471. The shape of microparticles and nanoparticles may be determined by Scanning electron microscopy. Spherically moieties is achieved. In some embodiments, the functional shaped nanoparticles are used in certain embodiments, for groups, which may be the same or different, terminal or circulation through the bloodstream. If desired, the particles internal, of each comonomer precursor comprise an amino, may be fabricated using known techniques into other shapes acid, imidazole, hydroxyl, thio, acyl halide, —HC—CH . that are more useful for a specific application. —C=C group, or derivative thereof. In some embodi 0472. In addition to intracellular delivery of a topoi ments, the two functional groups are the same and are located somerase inhibitor, it also possible that particles of the CDP at termini of the comonomer precursor. In some embodi topoisomerase inhibitor conjugates, such as microparticles or ments, a comonomer contains one or more pendant groups nanoparticles, may undergo endocytosis, thereby obtaining with at least one functional group through which reaction and access to the cell. The frequency of Such an endocytosis thus linkage of a therapeutic agent can be achieved. In some process will likely depend on the size of any particle. embodiments, the functional groups, which may be the same 0473. In one embodiment, the surface charge of the mol or different, terminal or internal, of each comonomer pendant ecule is neutral, or slightly negative. In some embodiments, group comprise an amino, acid, imidazole, hydroxyl, thiol, the Zeta potential of the particle surface is from about-80 mV acyl halide, ethylene, ethyne group, or derivative thereof. In to about 50 mV. Some embodiments, the pendant group is a Substituted or unsubstituted branched, cyclic or straight chain C1-C10 CDPs, Methods of Making Same, and Methods of Conjugat alkyl, orarylalkyl optionally containing one or more heteroa ing CDPs to Topoisomerase Inhibitors toms within the chain or ring. 0474 Generally, the CDP-topoisomerase inhibitor conju 0479. In some embodiments, the cyclodextrin moiety gates, particles and compositions described herein can be comprises an alpha, beta, or gamma cyclodextrin moiety. prepared in one of two ways: monomers bearing topoi 0480. In some embodiments, the CDP is suitable for the Somerase inhibitors, targeting ligands, and/or cyclodextrin attachment of Sufficient topoisomerase inhibitor Such that up moieties can be polymerized, or polymer backbones can be to at least 3%. 5%, 10%, 11%, 12%, 13%, 14%, 15%, 20%, derivatized with topoisomerase inhibitors, targeting ligands, 25%, 30%, or even 35% by weight of the CDP when conju and/or cyclodextrin moieties. Exemplary methods of making gated, is topoisomerase inhibitor. CDPs and CDP-topoisomerase inhibitor conjugates, particles 0481. In some embodiments, the CDP has a molecular and compositions are described, for example, in U.S. Pat. No. weight of 10,000-500,000 amu. In some embodiments, the 7,270.808, the contents of which is incorporated herein by cyclodextrin moieties make up at least about 2%. 5%, 10%, reference in its entirety. 20%, 30%, 50% or 80% of the CDP by weight. US 2011/O160 159 A1 Jun. 30, 2011 40

0482 In some embodiments, a CDP of the following for mula can be made by the scheme below:

OH

OH O O O OH HO HO O HO OH

O HO O O OH OH HO O O S OH NH NH-R S HO pi O OH HO O O OH

0483 wherein R is of the form:

O

O :

0484 comprising the steps of: 0485 reacting a compound of the formula below:

OH O OH O O OH HO O OH HO O O HO HO HO HO O OH

NH DO S O HO O 2

HN OH OH HO 5-O S O O O OH OH O

O O O OH HO US 2011/O160 159 A1 Jun. 30, 2011 41

0486 with a compound of the formula below:

O O O O --~~ LG --~~ LG 0489. In some embodiments, is

0487 wherein the group O O O O

N N

O O. -k Su-N iii. 0490. In some embodiments, the solvent is a polar aprotic solvent. In some embodiments, the solvent is DMSO. has a Mw of 3.4 kDa or less and n is at least four 0491. In some embodiments, the method also includes the s steps of dialysis; and lyophilization. 0488 in the presence of a non-nucleophilic organic base in 0492. In some embodiments, a CDP provided below can a solvent. be made by the following scheme:

OH

OH O O

OH HO HO O HO OH

HO

O OH HO PH OH O HO O O OH

wherein R is of the form: 0493 with a compound provided below:

N SN O N No lsO 1nue-n-r)So-1N1 O O NN O NN US 2011/O160 159 A1 Jun. 30, 2011 42 C --ro-continued wherein the group

has a Mw of 3.4 kDa; in the presence of a non-nucleophilic organic base in DMSO; 0494 and dialyzing and lyophilizing the following poly C

OH

O OH O O HO O O HHO OH

HO HO O HO OH O O S NH-R N OH HO S H pi

HO O O OH O PH OH O HO O

O O OH HO

0495. The present invention further contemplates CDPs and CDP-conjugates synthesized using CD-biscysteine monomer and a di-NHS ester Such as PEG-DiSPA or PEG BTC as shown in Scheme I.

Scheme I

O O O O DMSO HN NH2 -- N N - - S S 1 A No O O iii. O O HO O O OH Mw PEG = 3400 US 2011/O160 159 A1 Jun. 30, 2011

-continued EDCNHS -e- gly-CPT sc O r 95-98% Mn 55,700; Mwlec 99,500; Mw/Min = 1.74 - rs -

CPT CPT

Scheme XIII, as provided above, includes embodiments 0501. In some embodiments, the CDP is made by a pro where gly-CPT is absent in cess comprising: providing cyclodextrin moiety precursors, 0496 one or more positions as provided above. This can providing comonomer precursors, and copolymerizing said be achieved, for example, when less than 100% yield is cyclodextrin moiety precursors and comonomer precursors to achieved when coupling the CPT to the polymer and/or when thereby make a CDP comprising cyclodextrin moieties and less than an equivalent amount of CPT is used in the reaction. comonomers. In some embodiments, the CDP is conjugated Accordingly, the loading of the topoisomerase inhibitor Such with a topoisomerase inhibitor Such as camptothecin to pro as camptothecin, by weight of the polymer, can vary. There vide a CDP-topoisomerase inhibitor conjugate. fore, while Scheme XIII depicts CPT at each cysteine residue 0502. In some embodiments, the method includes provid of each polymer subunit, the CDP-CPT conjugate can have ing cyclodextrin moiety precursors modified to bear one reac less than 2 CPT molecules attached to any given polymer tive site at each of exactly two positions, and reacting the subunit of the CDP. For example, in one embodiment, the cyclodextrin moiety precursors with comonomer precursors CDP-CPT conjugate includes several polymer subunits and having exactly two reactive moieties capable of forming a each of the polymer Subunits can independently include two, covalent bond with the reactive sites under polymerization one or no CPT attached at each cysteine residue of the poly conditions that promote reaction of the reactive sites with the mer Subunit. In addition, the particles and compositions can reactive moieties to form covalent bonds between the include CDP-CPT conjugates having two, one or no CPT comonomers and the cyclodextrin moieties, whereby a CDP attached at each cysteine residue of each polymer Subunit of comprising alternating units of a cyclodextrin moiety and a the CDP-CPT conjugate and the conjugates include a mixture comonomer is produced. of CDP-CPT conjugates that can vary as to the number of 0503. In some embodiments, the topoisomerase inhibitor CPTs attached to the gly at each of the polymer subunits of the is attached to the CDP via a linker. In some embodiments, the conjugates in the particle or composition. linker is cleaved under biological conditions. 0497. In some embodiments, a CDP-topoisomerase 0504. In some embodiments, the topoisomerase inhibitor inhibitor conjugate can be made by providing a CDP com makes up at least 5%, 10%, 11%, 12%, 13%, 14%, 15%, 20%, prising cyclodextrin moieties and comonomers which do not 25%, 30%, or even 35% by weight of the CDP-topoisomerase contain cyclodextrin moieties (comonomers), wherein the inhibitor conjugate. cyclodextrin moieties and comonomers alternate in the CDP 0505. In some embodiments, the comonomer comprises and wherein the CDP comprises at least four cyclodextrin polyethylene glycol of molecular weight 3,400 Da, the cyclo moieties and at least four comonomers; and attaching a topoi dextrin moiety comprises beta-cyclodextrin, the theoretical somerase inhibitor to the CDP. maximum loading of camptothecin on a CDP-camptothecin 0498. In some embodiments, one or more of the topoi conjugate is 13%, and camptothecin is 6-10% by weight of somerase inhibitor moieties in the CDP-topoisomerase the CDP-camptothecin conjugate. inhibitor conjugate can be replaced with another therapeutic 0506. In some embodiments, the comonomer precursor is agent, e.g., another anticancer agent or anti-inflammatory a compound containing at least two functional groups agent. through which reaction and thus linkage of the cyclodextrin 0499. In some embodiments, the topoisomerase inhibitor moieties is achieved. In some embodiments, the functional is attached to the water soluble linear polymer via a linker. In groups, which may be the same or different, terminal or Some embodiments, the topoisomerase inhibitor is attached internal, of each comonomer precursor comprise an amino, to the water soluble linear polymer through an attachment that acid, imidazole, hydroxyl, thio, acyl halide, —HC—CH . is cleaved under biological conditions to release the topoi —C=C group, or derivative thereof. In some embodi Somerase inhibitor. In some embodiments, the topoisomerase ments, the two functional groups are the same and are located inhibitor is attached to the water soluble linear polymer at a at termini of the comonomer precursor. In some embodi cyclodextrin moiety or a comonomer. In some embodiments, ments, a comonomer contains one or more pendant groups the topoisomerase inhibitor is attached to the water soluble with at least one functional group through which reaction and linear polymer via an optional linker to a cyclodextrin moiety thus linkage of a therapeutic agent is achieved. In some O a COOOC. embodiments, the functional groups, which may be the same 0500. In some embodiments, the cyclodextrin moieties or different, terminal or internal, of each comonomer pendant comprise linkers to which therapeutic agents are linked group comprise an amino, acid, imidazole, hydroxyl, thiol, US 2011/O160 159 A1 Jun. 30, 2011 44 acyl halide, ethylene, ethyne group, or derivative thereof. In topoisomerase inhibitor over a period of 6 hours to a month. Some embodiments, the pendant group is a Substituted or In some embodiments, upon administration of the CDP-to unsubstituted branched, cyclic or straight chain C1-C10 poisomerase inhibitor conjugate, particle or composition to a alkyl, orarylalkyl optionally containing one or more heteroa subject the rate of topoisomerase inhibitor release is depen toms within the chain or ring. dent primarily upon the rate of hydrolysis as opposed to 0507. In some embodiments, the cyclodextrin moiety enzymatic cleavage. comprises an alpha, beta, or gamma cyclodextrin moiety. 0510. In some embodiments, the CDP-topoisomerase 0508. In some embodiments, the topoisomerase inhibitor is poorly soluble in water. inhibitor conjugate, particle or composition has a molecular 0509. In some embodiments, administration of the CDP weight of 10,000-500,000 amu. topoisomerase inhibitor conjugate, particle or composition to 0511. In some embodiments, the cyclodextrin moieties a subject results in release of the topoisomerase inhibitor over makeup at least about 2%. 5%, 10%, 20%, 30%, 50% or 80% a period of at least 6 hours. In some embodiments, adminis of the polymer by weight. tration of the CDP-topoisomerase inhibitor conjugate, par 0512. In some embodiments, a CDP-polymer conjugate of ticle or composition to a Subject results in release of the the following formula can be made as follows:

OH

O OH O O HO O OH HO OH

D HO HO O O HO L O OH O O S HO O N-1- t H OH S ^k O pi HO O O O OH O L PH OH O HO O

O O OH HO

providing a polymer below:

OH

O OH O O HO O O HHO OH

O O HO HO HO O OH O O O S O N OH HO N-). H S ^-k O pi HO O O O OH O OH OH OH O HO O

O O OH HO US 2011/O160 159 A1 Jun. 30, 2011 and coupling the polymer with a plurality of L-D moieties, wherein L is a linker, or absent and D is topoisomerase inhibi tor Such as camptothecin or a camptothecin derivative, to provide:

OH

O OH O O HO O OH HO

D HO HO O O HO L O OH O O S O N OH HO S ^-k N-) O HO O O pi O OH O L OH OH O HO O

O O OH HO wherein the group 0517. In certain instances, the CDPs are random copoly mers, in which the different subunits and/or other monomeric units are distributed randomly throughout the polymer chain. Thus, where the formula X, Y, Z appears, wherein X,Y --Su-N and Z are polymer Subunits, these subunits may be randomly interspersed throughout the polymer backbone. In part, the has a Mw of 3.4 kDa or less and n is at least 4, wherein on the term "random' is intended to refer to the situation in which final product, L can be a linker, a bond, or OH, and D can be the particular distribution or incorporation of monomeric a topoisomerase inhibitor (e.g., camptothecin or a camptoth units in a polymer that has more than one type of monomeric ecin derivative) or absent. units is not directed or controlled directly by the synthetic 0513. In some embodiments, one or more of the topoi protocol, but instead results from features inherent to the somerase inhibitor moieties in the CDP-topoisomerase polymer system, such as the reactivity, amounts of Subunits inhibitor conjugate can be replaced with another therapeutic and other characteristics of the synthetic reaction or other agent, e.g., another anticancer agent or anti-inflammatory methods of manufacture, processing, or treatment. agent. 0514. The reaction scheme as provided above includes Pharmaceutical Compositions embodiments where L-D is absent in one or more positions as provided above. This can be achieved, for example, when less 0518. In another aspect, the present invention provides a than 100% yield is achieved when coupling the topoi composition, e.g., a pharmaceutical composition, comprising somerase inhibitor-linker to the polymer and/or when less a CDP-topoisomerase inhibitor conjugate or particle and a than an equivalent amount of topoisomerase inhibitor-linker pharmaceutically acceptable carrier or adjuvant. is used in the reaction. Accordingly, the loading of the topoi 0519 In some embodiments, a pharmaceutical composi Somerase inhibitor, by weight of the polymer, can vary, for tion may include a pharmaceutically acceptable salt of a example, the loading of the topoisomerase inhibitor can be at compound described herein, e.g., a CDP-topoisomerase least about 3% by weight, e.g., at least about 5%, at least about inhibitor conjugate, particle or composition. Pharmaceuti 8%, at least about 10%, at least about 11%, at least about 12%, cally acceptable salts of the compounds described herein at least about 13%, at least about 14%, at least about 15%, or include those derived from pharmaceutically acceptable inor at least about 20%. ganic and organic acids and bases. Examples of suitable acid 0515. In some embodiments, at least a portion of the L salts include acetate, adipate, benzoate, benzenesulfonate, moieties of L-D is absent. In some embodiments, each L is butyrate, citrate, digluconate, dodecylsulfate, formate, fuma independently an amino acid or derivative thereof (e.g., gly rate, glycolate, hemisulfate, heptanoate, hexanoate, hydro cine). chloride, hydrobromide, hydroiodide, lactate, maleate, mal 0516. In some embodiments, the coupling of the polymer onate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, with the plurality of L-D moieties results in the formation of nitrate, palmoate, phosphate, picrate, pivalate, propionate, a plurality of amide bonds. salicylate, Succinate, Sulfate, tartrate, tosylate and unde US 2011/O160 159 A1 Jun. 30, 2011 46 canoate. Salts derived from appropriate bases include alkali Dehydrated Alcohol, USP and a nonionic surfactant, such as metal (e.g., sodium), alkaline earth metal (e.g., magnesium), a polyoxyethylated castor oil surfactant available from GAF ammonium and N-(alkyl) salts. This invention also envi Corporation, Mount Olive, N.J., under the trademark, Cre sions the quaternization of any basic nitrogen-containing mophor EL. In some embodiments, the lyophilized CDP groups of the compounds described herein. Water or oil topoisomerase inhibitor conjugate, particle or composition is soluble or dispersible products may be obtained by such reconstituted in water for infusion. The lyophilized product quaternization. and vehicle for reconstitution can be packaged separately in 0520 Wetting agents, emulsifiers and lubricants, such as appropriately light-protected vials, e.g., amber or other col Sodium lauryl Sulfate and magnesium Stearate, as well as ored vials. To minimize the amount of Surfactant in the recon coloring agents, release agents, coating agents, Sweetening, stituted Solution, only a Sufficient amount of the vehicle may flavoring and perfuming agents, preservatives and antioxi be provided to form a solution having a concentration of dants can also be present in the compositions. about 2 mg/mL to about 4 mg/mL of the CDP-topoisomerase 0521 Examples of pharmaceutically acceptable antioxi inhibitor conjugate, particle or composition. Once dissolution dants include: (1) water Soluble antioxidants, such as ascorbic of the drug is achieved, the resulting solution is further diluted acid, cysteine hydrochloride, Sodium bisulfate, Sodium met prior to injection with a suitable parenteral diluent. Such abisulfite, sodium sulfite and the like; (2) oil-soluble antioxi diluents are well known to those of ordinary skill in the art. dants, such as ascorbyl palmitate, butylated hydroxyanisole These diluents are generally available in clinical facilities. It (BHA), butylated hydroxytoluene (BHT), lecithin, propyl is, however, within the scope of the present invention to gailate, alpha-tocopherol, and the like; and (3) metal chelat package the Subject CDP-topoisomerase inhibitor conjugate, ing agents, such as citric acid, ethylenediamine tetraacetic particle or composition with a third vial containing Sufficient acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the parenteral diluent to prepare the final concentration for like. administration. A typical diluent is Lactated Ringer's Injec 0522. A composition may include a liquid used for sus tion. pending a CDP-topoisomerase inhibitor conjugate, particle 0527. The final dilution of the reconstituted CDP-topoi or composition, which may be any liquid Solution compatible Somerase inhibitor conjugate, particle or composition may be with the CDP-topoisomerase inhibitor conjugate, particle or carried out with other preparations having similar utility, for composition, which is also suitable to be used in pharmaceu example, 5% Dextrose Injection, Lactated Ringer's and Dex tical compositions, such as a pharmaceutically acceptable trose for Injection (D5W), Sterile Water for Injection, and the nontoxic liquid. Suitable suspending liquids including but are like. However, because of its narrow pH range, pH 6.0 to 7.5, not limited to Suspending liquids selected from the group Lactated Ringer's Injection is most typical. Per 100 mL. consisting of water, aqueous Sucrose syrups, corn Syrups, Lactated Ringer's Injection contains Sodium Chloride USP Sorbitol, polyethylene glycol, propylene glycol, and mixtures 0.6 g. Sodium Lactate 0.31 g, Potassium chloride USP 0.03 g thereof. and Calcium Chloride2H2O USP 0.02 g. The osmolarity is 0523. A composition described herein may also include 275 mOsmol/L, which is very close to isotonicity. another component, such as an antioxidant, antibacterial, 0528. The compositions may conveniently be presented in buffer, bulking agent, chelating agent, an inert gas, a tonicity unit dosage form and may be prepared by any methods well agent and/or a viscosity agent. known in the art of pharmacy. The dosage form can be, e.g., in 0524. In one embodiment, the CDP-topoisomerase inhibi a bog, e.g., a bag for infusion or intraperitoneal administra tor conjugate, particle or composition is provided in lyo tion. The amount of active ingredient which can be combined philized form and is reconstituted prior to administration to a with a carrier material to produce a single dosage form will subject. The lyophilized CDP-topoisomerase inhibitor con vary depending upon the host being treated, the particular jugate, particle or composition can be reconstituted by a mode of administration. The amount of active ingredient diluent Solution, such as a salt or saline Solution, e.g., a which can be combined with a carrier material to produce a sodium chloride solution having a pH between 6 and 9, lac single dosage form will generally be that amount of the com tated Ringer's injection solution, or a commercially available pound which produces a therapeutic effect. Generally, out of diluent, such as PLASMA-LYTEA Injection pH 7.4(R) (Bax one hundred percent, this amount will range from about 1 ter, Deerfield, Ill.). percent to about ninety-nine percent of active ingredient, 0525. In one embodiment, a lyophilized formulation preferably from about 5 percent to about 70 percent, most includes a lyoprotectant or stabilizer to maintain physical and preferably from about 10 percent to about 30 percent. chemical stability by protecting the CDP-topoisomerase inhibitor conjugate, particle or composition from damage Routes of Administration from crystal formation and the fusion process during freeze drying. The lyoprotectant or stabilizer can be one or more of 0529. The pharmaceutical compositions described herein polyethylene glycol (PEG), a PEG lipid conjugate (e.g., PEG may be administered orally, parenterally (e.g., via intrave ceramide or D-alpha-tocopheryl polyethylene glycol 1000 nous, Subcutaneous, intracutaneous, intramuscular, intraar succinate), poly(vinyl alcohol) (PVA), poly(vinylpyrroli ticular, intraarterial, intraperitoneal, intrasynovial, intraster done) (PVP), polyoxyethylene esters, poloxomers, Tweens, nal, intrathecal, intralesional or intracranial injection), lecithins, saccharides, oligosaccharides, polysaccharides and topically, mucosally (e.g., rectally or vaginally), nasally, buc polyols (e.g., trehalose, mannitol, Sorbitol, lactose. Sucrose, cally, ophthalmically, via inhalation spray (e.g., delivered via glucose and dextran), salts and crown ethers. In one embodi nebulzation, propellant or a dry powder device) or via an ment, the lyoprotectant is mannitol. implanted reservoir. Typically, the compositions are in the 0526 In some embodiments, the lyophilized CDP-topoi form of injectable or infusible solutions. The preferred mode Somerase inhibitor conjugate, particle or composition is of administration is, e.g., intravenous, Subcutaneous, intrap reconstituted with a mixture of equal parts by volume of eritoneal, intramuscular. US 2011/O160 159 A1 Jun. 30, 2011 47

0530 Pharmaceutical compositions suitable for 0537 Tablets, and other solid dosage forms, such as dra parenteral administration comprise gees, capsules, pills and granules, may optionally be scored or 0531 one or more CDP-topoisomerase inhibitor conju prepared with coatings and shells, such as enteric coatings gate(s), particle(s) or composition(s) in combination with one and other coatings well known in the pharmaceutical-formu or more pharmaceutically acceptable sterile isotonic aqueous lating art. They may also be formulated so as to provide slow or nonaqueous Solutions, dispersions, Suspensions or emul or controlled release of the active ingredient therein using, for sions, or sterile powders which may be reconstituted into example, hydroxypropylmethyl cellulose in varying propor sterile injectable solutions or dispersions just prior to use, tions to provide the desired release profile, other polymer which may contain antioxidants, buffers, bacteriostats, Sol matrices, liposomes and/or microspheres. They may be ster utes which render the formulation isotonic with the blood of ilized by, for example, filtration through a bacteria-retaining the intended recipient or Suspending or thickening agents. filter, or by incorporating sterilizing agents in the form of 0532. Examples of Suitable aqueous and nonaqueous car riers which may be employed in the pharmaceutical compo sterile solid compositions which can be dissolved in sterile sitions include water, ethanol, polyols (such as glycerol, pro water, or some other sterile injectable medium immediately pylene glycol, polyethylene glycol, and the like), and Suitable before use. These compositions may also optionally contain mixtures thereof, vegetable oils, such as olive oil, and inject opacifying agents and may be of a composition that they able organic esters, such as ethyl oleate. Proper fluidity can be release the active ingredient(s) only, or preferentially, in a maintained, for example, by the use of coating materials, such certain portion of the gastrointestinal tract, optionally, in a as lecithin, by the maintenance of the required particle size in delayed manner. Examples of embedding compositions the case of dispersions, and by the use of Surfactants. which can be used include polymeric Substances and waxes. 0533. These compositions may also contain adjuvants The active ingredient can also be in micro-encapsulated form, Such as preservatives, wetting agents, emulsifying agents and if appropriate, with one or more of the above-described dispersing agents. Prevention of the action of microorgan excipients. isms may be ensured by the inclusion of various antibacterial 0538 Liquid dosage forms for oral administration include and agents, for example, paraben, chlorobutanol, pharmaceutically acceptable emulsions, microemulsions, phenol sorbic acid, and the like. It may also be desirable to Solutions, Suspensions, syrups and elixirs. In addition to the include isotonic agents. Such as , sodium chloride, and CDP-topoisomerase inhibitor conjugate, particle or compo the like into the compositions. In addition, prolonged absorp sition, the liquid dosage forms may contain inert diluents tion of the injectable pharmaceutical form may be brought commonly used in the art, Such as, for example, water or other about by the inclusion of agents which delay absorption Such Solvents, Solubilizing agents and emulsifiers, such as ethyl as aluminum monostearate and gelatin. alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, 0534. In some cases, in order to prolong the effect of a benzyl alcohol, benzyl benzoate, propylene glycol. 1,3-buty drug, it is desirable to slow the absorption of the agent from lene glycol, oils (in particular, cottonseed, groundnut, corn, Subcutaneous or intramuscular injection. This may be accom germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl plished by the use of a liquid Suspension of crystalline or alcohol, polyethylene glycols andfatty acid esters of sorbitan, amorphous material having poor water solubility. The rate of and mixtures thereof. absorption of the CDP-topoisomerase inhibitor conjugate, 0539 Besides inert diluents, the oral compositions can particle or composition then depends upon its rate of disso also include adjuvants such as wetting agents, emulsifying lution which, in turn, may depend upon crystal size and crys and Suspending agents, Sweetening, flavoring, coloring, per talline form. Alternatively, delayed absorption of a parenter fuming and preservative agents. ally administered drug form is accomplished by dissolving or 0540 Suspensions, in addition to the CDP-topoisomerase Suspending the CDP-topoisomerase inhibitor conjugate, par inhibitor conjugate, particle or composition may contain Sus ticle or composition in an oil vehicle. pending agents as, for example, ethoxylated isostearyl alco 0535 Pharmaceutical compositions suitable for oral hols, polyoxyethylene Sorbitol and Sorbitan esters, microc administration may be in the form of capsules, cachets, pills, rystalline cellulose, aluminum metahydroxide, bentonite, tablets, gums, lozenges (using a flavored basis, usually agar-agar and tragacanth, and mixtures thereof. Sucrose and acacia or tragacanth), powders, granules, or as a 0541. Pharmaceutical compositions suitable for topical Solution or a suspension in an aqueous or non-aqueous liquid, administration are useful when the desired treatment involves or as an oil-in-water or water-in-oil liquid emulsion, or as an areas or organs readily accessible by topical application. For elixir or syrup, or as pastilles (using an inert base, such as application topically to the skin, the pharmaceutical compo gelatin and glycerin, or Sucrose and acacia) and/or as mouth sition should beformulated with a suitable ointment contain washes and the like, each containing a predetermined amount ing the active components Suspended or dissolved in a carrier. of an agent as an active ingredient. A compound may also be Carriers for topical administration of the aparticle described administered as a bolus, electuary or paste. herein include, but are not limited to, mineral oil, liquid 0536. A tablet may be made by compression or molding, petroleum, white petroleum, propylene glycol, polyoxyeth optionally with one or more accessory ingredients. Com ylene polyoxypropylene compound, emulsifying wax and pressed tablets may be prepared using binder (for example, water. Alternatively, the pharmaceutical composition can be gelatin or hydroxypropylmethyl cellulose), lubricant, inert formulated with a suitable lotion or cream containing the diluent, preservative, disintegrant (for example, sodium active particle Suspended or dissolved in a carrier with Suit starch glycolate or cross-linked sodium carboxymethyl cel able emulsifying agents. Suitable carriers include, but are not lulose), Surface-active or dispersing agent. Molded tablets limited to, mineral oil, Sorbitan monostearate, polysorbate 60, may be made by molding in a Suitable machine a mixture of cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl the powdered peptide or peptidomimetic moistened with an alcohol and water. The pharmaceutical compositions inert liquid diluent. described herein may also be topically applied to the lower US 2011/O160 159 A1 Jun. 30, 2011 48 intestinal tract by rectal Suppository formulation or in a Suit the agent is administered. Preferably the dose of the CDP able enema formulation. Topically-transdermal patches are topoisomerase inhibitor conjugate, particle or composition is also included herein. a dose described herein. 0542. The pharmaceutical compositions described herein 0550. In one embodiment, the subject receives 1, 2, 3, up may be administered by nasal aerosol or inhalation. Such to 10 treatments, or more, or until the disorder or a symptom compositions are prepared according to techniques well of the disorder is cured, healed, alleviated, relieved, altered, remedied, ameliorated, palliated, improved or affected. For known in the art of pharmaceutical formulation and may be example, the Subject receives an infusion once every 1, 2, 3 or prepared as Solutions in Saline, employing benzyl alcohol or 4 weeks until the disorder or a symptom of the disorder is other Suitable preservatives, absorption promoters to enhance cured, healed, alleviated, relieved, altered, remedied, amelio bioavailability, fluorocarbons, and/or other solubilizing or rated, palliated, improved or affected. Preferably, the dosing dispersing agents known in the art. schedule is a dosing schedule described herein. 0543. The pharmaceutical compositions described herein 0551. The CDP-topoisomerase inhibitor conjugate, par may also be administered in the form of Suppositories for ticle or composition can be administered as a first line therapy, rectal or vaginal administration. Suppositories may be pre e.g., alone or in combination with an additional agent or pared by mixing one or more CDP-topoisomerase inhibitor agents. In other embodiments, a CDP-topoisomerase inhibi conjugate, particle or composition described herein with one tor conjugate, particle or composition is administered after a or more Suitable non-irritating excipients which is solid at Subject has developed resistance to, has failed to respond to or room temperature, but liquid at body temperature. The com has relapsed after a first line therapy. The CDP-topoisomerase position will therefore melt in the rectum or vaginal cavity inhibitor conjugate, particle or composition can be adminis and release the CDP-topoisomerase inhibitor conjugate, par tered in combination with a second agent. Preferably, the ticle or composition. Such materials include, for example, CDP-topoisomerase inhibitor conjugate, particle or compo cocoa butter, polyethylene glycol, a Suppository wax or a sition is administered in combination with a second agent salicylate. Compositions of the present invention which are described herein. Suitable for vaginal administration also include pessaries, 0552 Kits tampons, creams, gels, pastes, foams or spray formulations 0553 A CDP-topoisomerase inhibitor conjugate, particle containing Such carriers as are known in the art to be appro or composition described herein may be provided in a kit. The priate. kit includes a CDP-topoisomerase inhibitor conjugate, par 0544 Ophthalmic formulations, eye ointments, powders, ticle or composition described herein and, optionally, a con Solutions and the like, are also contemplated as being within tainer, a pharmaceutically acceptable carrier and/or informa the scope of the invention. tional material. The informational material can be descriptive, instructional, marketing or other material that 0545 Dosages and Dosing Regimens relates to the methods described herein and/or the use of the 0546. The CDP-topoisomerase inhibitor conjugate, par CDP-topoisomerase inhibitor conjugate, particle or compo ticle or composition can be formulated into pharmaceutically sition for the methods described herein. acceptable dosage forms by conventional methods known to 0554. The informational material of the kits is not limited those of skill in the art. in its form. In one embodiment, the informational material 0547 Actual dosage levels of the active ingredients in the can include information about production of the CDP-topoi pharmaceutical compositions of this invention may be varied Somerase inhibitor conjugate, particle or composition, physi So as to obtain an amount of the active ingredient which is cal properties of the CDP-topoisomerase inhibitor conjugate, effective to achieve the desired therapeutic response for a particle or composition, concentration, date of expiration, particular subject, composition, and mode of administration, batch or production site information, and so forth. In one without being toxic to the subject. embodiment, the informational material relates to methods 0548. In one embodiment, the CDP-topoisomerase inhibi for administering the CDP-topoisomerase inhibitor conju tor conjugate, particle or composition is administered to a gate, particle or composition, e.g., by a route of administra subject at a dosage of, e.g., about 1 to 40 mg/m, about 3 to 35 tion described herein and/or at a dose and/or dosing schedule mg/m, about 9 to 40 mg/m, e.g., about 1,3,5,6,7,8,9, 10, described herein. 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 0555. In one embodiment, the informational material can 28, 29, 30, 31, 32,33,34, 35, 36, 37,38, 39, 40 mg/m of the include instructions to administer a CDP-topoisomerase topoisomerase inhibitor. Administration can be at regular inhibitor conjugate, particle or composition described herein intervals, such as weekly, or every 2, 3, 4, 5 or 6 weeks. The in a suitable manner to perform the methods described herein, administration can be over a period of from about 10 minutes e.g., in a Suitable dose, dosage form, or mode of administra to about 6 hours, e.g., from about 30 minutes to about 2 hours, tion (e.g., a dose, dosage form, or mode of administration from about 45 minutes to 90 minutes, e.g., about 30 minutes, described herein). In another embodiment, the informational 45 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours or more. material can include instructions to administer a CDP-topoi The CDP-topoisomerase inhibitor conjugate, particle or com Somerase inhibitor conjugate, particle or composition position can be administered, e.g., by intravenous or intrap described herein to a Suitable subject, e.g., a human, e.g., a eritoneal administration. human having or at risk for a disorder described herein. In 0549. In one embodiment, the CDP-topoisomerase inhibi another embodiment, the informational material can include tor conjugate, particle or composition is administered as a instructions to reconstitute a CDP-topoisomerase inhibitor bolus infusion or intravenous push, e.g., over a period of 15 conjugate, particle or composition described herein into a minutes, 10 minutes, 5 minutes or less. In one embodiment, pharmaceutically acceptable composition. the CDP-topoisomerase inhibitor conjugate, particle or com 0556. In one embodiment, the kit includes instructions to position is administered in an amount Such the desired dose of use the CDP-topoisomerase inhibitor conjugate, particle or US 2011/O160 159 A1 Jun. 30, 2011 49 composition, Such as for treatment of a subject. The instruc for using a CDP-topoisomerase inhibitor conjugate, particle tions can include methods for reconstituting or diluting the or composition described herein together with the other CDP-topoisomerase inhibitor conjugate, particle or compo ingredients. For example, the kit can include an agent which sition for use with a particular subject or in combination with reduces or inhibits one or more symptom of hypersensitivity, a particular chemotherapeutic agent. The instructions can a polysaccharide, and/or an agent which increases urinary also include methods for reconstituting or diluting the CDP excretion and/or neutralizes topoisomerase inhibitor conjugate, particle or composition 0560 one or more urinary metabolite. for use with a particular means of administration, such as by 0561. In another embodiment, the kit includes a second intravenous infusion or intraperitoneal administration. therapeutic agent, such as a second chemotherapeutic agent, 0557. In another embodiment, the kit includes instructions e.g., a chemotherapeutic agent or combination of chemo for treating a subject with a particular indication, Such as a therapeutic agents described herein. In one embodiment, the particular cancer, or a cancer at a particular stage. For second agent is in lyophilized or in liquid form. In one example, the instructions can be for a cancer or cancer at stage embodiment, the CDP-topoisomerase inhibitor conjugate, described herein, e.g., lung cancer (e.g., non Small cell lung particle or composition and the second therapeutic agent are cancer and/or small cell lung cancer, e.g., squamous cell in separate containers, and in another embodiment, the CDP non-Small cell and/or Small cell lung cancer) or ovarian can topoisomerase inhibitor conjugate, particle or composition cer. The instructions may also address first line treatment of a and the second therapeutic agent are packaged in the same Subject who has a particular cancer, or cancer at a stage container. described herein. The instructions can also address treatment 0562. In some embodiments, a component of the kit is of a subject who has been non-responsive to a first line stored in a sealed vial, e.g., with a rubber or silicone closure therapy or has become sensitive (e.g., has one or more unac (e.g., a polybutadiene or polyisoprene closure). In some ceptable side effect) to a first line therapy, such as a taxane, an embodiments, a component of the kit is stored under inert anthracycline, an antimetabolite, a Vinca alkaloid, a vascular conditions (e.g., under Nitrogen or another inert gas Such as endothelial growth factor (VEGF) pathway inhibitor, an epi Argon). In some embodiments, a component of the kit is dermal growth factor (EGF) pathway inhibitor, an alkylating stored under anhydrous conditions (e.g., with a desiccant). In agent, a platinum-based agent, a Vinca alkaloid. In another Some embodiments, a component of the kit is stored in a light embodiment, the instructions will describe treatment of blocking container Such as an amber vial. selected subjects with the CDP-topoisomerase inhibitor con 0563 A CDP-topoisomerase inhibitor conjugate, particle jugate, particle or composition. For example, the instructions or composition described herein can be provided in any form, can describe treatment of one or more of a Subject having a e.g., liquid, frozen, dried or lyophilized form. It is preferred cancer that has increased levels of KRAS and/or ST expres that a composition including the conjugate, particle or com Sion, e.g., as compared to a reference standard. position, e.g., a composition comprising a particle or particles 0558. The informational material of the kits is not limited that include a conjugate described herein be substantially in its form. In many cases, the informational material, e.g., pure and/or sterile. When a CDP-topoisomerase inhibitor instructions, is provided in printed matter, e.g., a printed text, conjugate, particle or composition described herein is pro drawing, and/or photograph, e.g., a label or printed sheet. vided in a liquid solution, the liquid solution preferably is an However, the informational material can also be provided in aqueous solution, with a sterile aqueous solution being pre other formats, such as Braille, computer readable material, ferred. In one embodiment, the CDP-topoisomerase inhibitor Video recording, or audio recording. In another embodiment, conjugate, particle or composition is provided in lyophilized the informational material of the kit is contact information, form and, optionally, a diluent Solution is provided for recon e.g., a physical address, email address, website, or telephone stituting the lyophilized agent. The diluent can include for number, where a user of the kit can obtain substantive infor example, a salt or saline solution, e.g., a Sodium chloride mation about a CDP-topoisomerase inhibitor conjugate, par solution having a pH between 6 and 9, lactated Ringer's ticle or composition described herein and/or its use in the injection solution, D5W, or PLASMA-LYTEA Injection pH methods described herein. The informational material can 7.4(R) (Baxter, Deerfield, Ill.). also be provided in any combination of formats. 0564. The kit can include one or more containers for the 0559. In addition to a CDP-topoisomerase inhibitor con composition containing a CDP-topoisomerase inhibitor con jugate, particle or composition described herein, the compo jugate, particle or composition described herein. In some sition of the kit can include other ingredients, such as a embodiments, the kit contains separate containers, dividers or Surfactant, a lyoprotectant or stabilizer, an antioxidant, an compartments for the composition and informational mate antibacterial agent, a bulking agent, a chelating agent, an inert rial. For example, the composition can be contained in a gas, a tonicity agent and/or a viscosity agent, a solvent or bottle, vial. IV admixture bag, IV infusion set, piggyback set buffer, a stabilizer, a preservative, a flavoring agent (e.g., a or syringe, and the informational material can be contained in bitter antagonist or a Sweetener), a fragrance, a dye or color a plastic sleeve or packet. In other embodiments, the separate ing agent, for example, to tint or color one or more compo elements of the kit are contained within a single, undivided nents in the kit, or other cosmetic ingredient, a pharmaceuti container. For example, the composition is contained in a cally acceptable carrier and/or a second agent for treating a bottle, vial or syringe that has attached thereto the informa condition or disorder described herein. Alternatively, the tional material in the form of a label. In some embodiments, other ingredients can be included in the kit, but in different the kit includes a plurality (e.g., a pack) of individual con compositions or containers than a CDP-topoisomerase tainers, each containing one or more unit dosage forms (e.g., inhibitor conjugate, particle or composition described herein. a dosage form described herein) of a CDP-topoisomerase In Such embodiments, the kit can include instructions for inhibitor conjugate, particle or composition described herein. admixing a CDP-topoisomerase inhibitor conjugate, particle For example, the kit includes a plurality of syringes, ampules, or composition described herein and the other ingredients, or foil packets, or blister packs, each containing a single unit US 2011/O160 159 A1 Jun. 30, 2011 50 dose of a particle described herein. The containers of the kits 0569. In some embodiments, the CDP-topoisomerase can be air tight, waterproof (e.g., impermeable to changes in inhibitor conjugate, particle or composition is administered moisture or evaporation), and/or light-tight. with at least one additional therapeutic agent, Such as a che 0565. The kit optionally includes a device suitable for motherapeutic agent. In certain embodiments, the CDP-to administration of the composition, e.g., a Syringe, inhalant, poisomerase inhibitor conjugate, particle or composition is pipette, forceps, measured spoon, dropper (e.g., eye dropper), administered in combination with one or more additional Swab (e.g., a cotton Swab or wooden Swab), or any Such chemotherapeutic agent, e.g., with one or more chemothera delivery device. In one embodiment, the device is a medical peutic agents described herein. Exemplary classes of chemo implant device, e.g., packaged for Surgical insertion. therapeutic agents include, e.g., the following: 0570 alkylating agents (including, without limitation, Combination Therapy nitrogen mustards, ethylenimine derivatives, alkylsulfonates, and ): mustard (Aminouracil 0566. The CDP-topoisomerase inhibitor conjugate, par Mustard R. Chlorethaminacil(R), Demethyldopan R. Desm ticle or composition may be used in combination with other ethyldopan R, Haemanthamine(R), Nordopan R. Uracil nitro known therapies. Administered "in combination', as used gen Mustard(R), Uracillost(R), UracilmostazaR), Uramustin R, herein, means that two (or more) different treatments are (R), (MustargenR), cyclophospha delivered to the subject during the course of the subject's mide (Cytoxan R, Neosar(R), Clafen R. Endoxan R, Procy affliction with the disorder, e.g., the two or more treatments toxR), Revimmune), ifosfamide (MitoxanaR), melphalan are delivered after the subject has been diagnosed with the (Alkeran R), (Leukeran R), disorder and before the disorder has been cured or eliminated (Amedel(R), VercyteR), (Hemel(R), or treatment has ceased for other reasons. In some embodi Hexylen R, Hexastat(R), triethylenethiophosphoramine, ments, the delivery of one treatment is still occurring when Temozolomide (Temodar R), (Thioplex(R), the delivery of the second begins, so that there is overlap in (Busilvex(R), Myleran R), (BiCNUR), terms of administration. This is sometimes referred to herein (CeeNUR), streptozocin (Zanosar R), and as “simultaneous” or “concurrent delivery’. In other embodi (DTIC-Dome(R). ments, the delivery of one treatment ends before the delivery (0571 anti-EGFR antibodies (e.g., cetuximab (Erbitux(R) of the other treatment begins. In some embodiments of either and panitumumab (Vectibix(R). case, the treatment is more effective because of combined 0572 anti-HER-2 antibodies (e.g., trastuzumab (Hercep administration. For example, the second treatment is more tin(R). effective, e.g., an equivalent effect is seen with less of the 0573 (including, without limitation, folic second treatment, or the second treatment reduces symptoms acid antagonists (also referred to herein as ), pyri to a greater extent, than would be seen if the second treatment midine analogs, analogs and deaminase were administered in the absence of the first treatment, or the inhibitors): (Rheumatrex(R), Trexall(R), 5-fluo analogous situation is seen with the first treatment. In some rouracil (Adrucil(R), Efudex(R, Fluoroplex(R), floxuridine embodiments, delivery is such that the reduction in a symp (FUDF(R), (Cytosar-UR, Tarabine PFS), 6-mer tom, or other parameter related to the disorder is greater than captopurine (Puri-NetholR)), 6-thioguanine (Thioguanine what would be observed with one treatment delivered in the TabloidR), phosphate (Fludara(R), absence of the other. The effect of the two treatments can be (Nipent(R), pemetrexed (AlimtaR), raltitrexed (Tomudex(R), partially additive, wholly additive, or greater than additive. (LeustatinR), (Clofarex(R), Clolar R), The delivery can be such that an effect of the first treatment (Puri-NetholR), capecitabine (Xeloda(R), delivered is still detectable when the second is delivered. (Arranon(R), (VidazaR) and gemcitab 0567 The CDP-topoisomerase inhibitor conjugate, par ine (Gemzar(R). Preferred antimetabolites include, e.g., ticle or composition and the at least one additional therapeutic 5- (Adrucil(R), Efudex(R, Fluoroplex(R), floxuri agent can be administered simultaneously, in the same or in dine (FUDF(R), capecitabine (Xeloda(R), pemetrexed (Al separate compositions, or sequentially. For sequential admin imtaR), raltitrexed (Tomudex(R) and gemcitabine istration, the CDP-topoisomerase inhibitor conjugate, par (Gemzar(R). ticle or composition can be administered first, and the addi (0574 vinca alkaloids: vinblastine (Velban(R), Velsar R), tional agent can be administered second, or the order of Vincristine (Vincasar R, OncovinR), vindesine (Eldisine(R). administration can be reversed. vinorelbine (Navelbine(R). 0568. In some embodiments, the CDP-topoisomerase 0575 platinum-based agents: carboplatin (Paraplater), inhibitor conjugate, particle or composition is administered ParaplatinR), cisplatin (PlatinolR), oxaliplatin (EloxatinR). in combination with other therapeutic treatment modalities, 0576 : daunorubicin (Cerubidine(R), Rubi including Surgery, radiation, cryoSurgery, and/or thermo domycin R.), doxorubicin (Adriamycin R), epirubicin (El therapy. Such combination therapies may advantageously uti lence(R), idarubicin (Idamycin R.), mitoxantrone (No lize lower dosages of the administered agent and/or other Vantrone(R), valrubicin (Valstar R). Preferred anthracyclines chemotherapeutic agent, thus avoiding possible toxicities or include daunorubicin (Cerubidine(R), Rubidomycin R) and complications associated with the various monotherapies. doxorubicin (Adriamycin R). The phrase “radiation' includes, but is not limited to, exter 0577 topoisomerase inhibitors: topotecan (Hycamtin R), nal-beam therapy which involves three dimensional, confor irinotecan (Camptosar(R), etoposide (Toposar R, VePesidr), mal radiation therapy where the field of radiation is designed (VumonR), lamellarin D, SN-38, camptothecin. to conform to the volume of tissue treated; interstitial-radia 0578 : paclitaxel (Taxol.R.), docetaxel (Taxo tion therapy where seeds of radioactive compounds are tere?R), larotaxel, cabazitaxel. implanted using ultrasound guidance; and a combination of 0579 : , epothilone B, epothilone external-beam therapy and interstitial-radiation therapy. D, BMS310705, dehydelone, ZK-Epothilone (ZK-EPO). US 2011/O160 159 A1 Jun. 30, 2011

0580 poly ADP-ribose polymerase (PARP) inhibitors: vartis), Lucentis (Ranibizumab) (Genentech/Novartis), (e.g., BSI 201, Olaparib (AZD-2281), ABT-888, AG014699, LY317615 (Enzastaurin) (Eli Lilly and Company), Macugen CEP 9722, MK 4827, KU-0059436 (AZD2281), LT-673, (Pegaptainib) (Pfizer), MEDI522 (Abegrin) (MedImmune), 3-aminobenzamide). MLN518 (Tandutinib) (Millennium), Neovastat (AE941/ 0581 : actinomycin (CosmegenR), Benefin) (Aeterna Zentaris). Nexavar (Bayer/Onyx), NM-3 (Blenoxane(R), hydroxyurea (DroxiaR), HydreaR), mitomy (Genzyme Corporation), NoScapine (Cougar Biotechnol cin (MitoZytrex(R, Mutamycin R). ogy), NPI-2358 (Nereus Pharmaceuticals), OSI-930 (OSI), 0582 immunomodulators: (Revlimid(R). Palomid 529 (Paloma Pharmaceuticals, Inc.), Panzem Cap thalidomide (Thalomid(R). sules (2ME2) (EntreMed), Panzem. NCD (2ME2) (En 0583 immune cellantibodies: alemtuzamab (Campath(R), treMed), PF-02341066 (Pfizer), PF-04554878 (Pfizer), PI-88 gemtuzumab (Myelotargr), rituximab (Rituxan R), to situ (Progen Industries/Medigen Biotechnology), PKC412 (No momab (Bexxar R). vartis), Polyphenon E (Green Tea Extract) (Polypheno E 0584 interferons (e.g., IFN-alpha (Alferon(R), Roferon International, Inc), PPI-2458 (Praecis Pharmaceuticals), AR), Intron(R-A) or IFN-gamma (ActimmuneR)). PTC299 (PTC 0585 interleukins: IL-1, IL-2 (Proleukin R), IL-24, IL-6 0588. Therapeutics), PTK787 (Vatalanib) (Novartis), (Sigosix(R), IL-12. PXD101 (Belinostat) (CuraGen Corporation), RAD001 0586 HSP90 inhibitors (e.g., geldanamycin or any of its (Everolimus) (Novartis), RAF265 (Novartis), Regorafenib derivatives). In certain embodiments, the HSP90 inhibitor is (BAY73-4506) (Bayer), Revlimid (Celgene), Retaane (Alcon selected from geldanamycin, 17-alkylamino-17-desmethox Research), SN38 (Liposomal) (Neopharm), SNS-032 (BMS 387032) (Sunesis), SOM230 (Pasireotide) (Novartis), ygeldanamycin (“17-AAG”) or 17-(2-dimethylaminoethyl) Squalamine (Genaera), Suramin, Sutent (Pfizer), Tarceva amino-17-desmethoxygeldanamycin (“17-DMAG”). (Genentech), TB-403 (Thrombogenics), Tempostatin (Col 0587 angiogenesis inhibitors which include, without lard Biopharmaceuticals), Tetrathiomolybdate (Sigma-Ald limitation A6 (Angstrom Pharmacueticals), ABT-510 (Ab rich), TG100801 (TargeCien), Thalidomide (Celgene Corpo bott Laboratories), ABT-627 (Atrasentan) (Abbott Laborato ration), Tinzaparin Sodium, TKI258 (Novartis), TRC093 ries/Xinlay), ABT-869 (Abbott Laboratories), Actimid (Tracon Pharmaceuticals Inc.), VEGF Trap (Aflibercept) (CC4047, ) (Celgene Corporation), (Regeneron Pharmaceuticals), VEGF Trap-Eye (Regeneron AdGVPEDF.11D (GenVec), ADH-1 (Exherin) (Adherex Pharmaceuticals), Veglin (VascGene Therapeutics), Bort Technologies), AEE788 (Novartis), AG-013736 (Axitinib) (Pfizer), AG3340 (Prinomastat) (Agouron Pharmaceuticals), eZomib (Millennium), XL184 (Exelixis), XL647 (Exelixis), AGX1053 (AngioGenex), AGX51 (AngioGenex), ALN-VSP XL784 (Exelixis), XL820 (Exelixis), XL999 (Exelixis), (ALN-VSPO2) (Alnylam Pharmaceuticals), AMG 386 (Am ZD6474 (AstraZeneca), Vorinostat (Merck), and ZSTK474. gen), AMG706 (Amgen), Apatinib (YN968D1) (Jiangsu 0589 anti-androgens which include, without limitation Hengrui Medicine), AP23573 (Ridaforolimus/MK8669) nilutamide (Nilandron(R) and bicalutamide (Caxodex(R). (Ariad Pharmaceuticals), AQ4N (Novavea), ARQ 197 (Ar 0590 antiestrogens which include, without limitation Qule), ASA404 (Novartis/Antisoma), Atiprimod (Callisto tamoxifen (Nolvadex(R), toremifene (Fareston(R), letrozole Pharmaceuticals), ATN-161 (Attenuon), AV-412 (Aveo Phar (Femara(R), (TeslacR), anastrozole (Arimi maceuticals), AV-951 (Aveo Pharmaceuticals), Avastin (Be dex(R), bicalutamide (Casodex(R), exemestane (Aromasin(R), vacizumab) (Genentech), AZD2171 (Cediranib/Recentin) flutamide (Eulexin R), fulvestrant (Faslodex(R), raloxifene (AstraZeneca), BAY 57-9352 (Telatinib) (Bayer), BEZ235 (EvistaR) Keoxifene?R) and raloxifene hydrochloride. (Novartis), BIBF1120 (Boehringer Ingelheim Pharmaceuti 0591 anti-hypercalcaemia agents which include without cals), BIBW 2992 (Boehringer Ingelheim Pharmaceuticals), limitation gallium (III) nitrate hydrate (GaniteR) and pam BMS-275291 (Bristol-Myers Squibb), BMS-582664 (Bri idronate disodium (ArediaR). vanib) (Bristol-Myers Squibb), BMS-690514 (Bristol-Myers 0592 inducers which include without limitation Squibb), Calcitriol, CCI-779 (Torisel) (Wyeth), CDP-791 ethanol, 2-3-(2,3-dichlorophenoxy)propylamino-(9Cl), (ImClone Systems), Ceflatonin (Homoharringtonine/HHT) gambogic acid, embelin and (TrisenoX(R). (ChemGenex Therapeutics), Celebrex (Celecoxib) (Pfizer), 0593. Aurora kinase inhibitors which include without CEP-7055 (Cephalon/Sanofi), CHIR-265 (Chiron Corpora limitation binucleine 2. tion), NGR-TNF, COL-3 (Metastat) (Collagenex Pharmaceu 0594 Bruton's tyrosine kinase inhibitors which include ticals), Combretastatin (Oxigene), CP-751,871 (Figitu without limitation terreic acid. mumab) (Pfizer), CP-547,632 (Pfizer), CS-7017 (Daiichi 0595 calcineurin inhibitors which include without limita Sankyo Pharma), CT-322 (Angiocept) (Adnexus), Curcumin, tion cypermethrin, deltamethrin, fenvalerate and tyrphostin 8. Dalteparin (Fragmin) (Pfizer), Disulfuram (Antabuse), 0596 CaM kinase II inhibitors which include without E7820 (Eisai Limited), E7080 (Eisai Limited), EMD 121974 limitation 5-Isoquinolinesulfonic acid, 4-(2S)-2-(5-iso (Cilengitide) (EMD Pharmaceuticals), ENMD-1198 (En quinolinylsulfonyl)methylaminol-3-oxo-3-4-phenyl-1-pip treMed), ENMD-2076 (EntreMed), Endostar (Simcere), erazinyl)propylphenyl ester and benzenesulfonamide Erbitux (ImClone/Bristol-Myers Squibb), EZN-2208 (Enzon 0597 CD45 tyrosine phosphatase inhibitors which Pharmaceuticals), EZN-2968 (Enzon Pharmaceuticals), include without limitation phosphonic acid. GC1008 (Genzyme), Genistein, GSK1363089 (Foretinib) 0598 CDC25 phosphatase inhibitors which include with (GlaxoSmithKline), GW786034 (Pazopanib) (GlaxoSmith out limitation 1.4-naphthalene dione, 2,3-bis(2-hydroxy Kline), GT-111 (Vascular Biogenics Ltd.), IMC-1 121 B (Ra ethyl)thio)-(9Cl). mucirumab) (ImClone Systems), IMC-18F 1 (ImClone Sys 0599 CHK kinase inhibitors which include without limi tems), IMC-3G3 (ImClone LLC), INCB007839 (Incyte tation debromohymenialdisine. Corporation), INGN 241 (Introgen Therapeutics), Iressa 0600 cyclooxygenase inhibitors which include without (ZD1839/Gefitinib), LBH589 (Faridak/Panobinostst) (No limitation 1H-indole-3-acetamide, 1-(4-chlorobenzoyl)-5- US 2011/O160 159 A1 Jun. 30, 2011 52 methoxy-2-methyl-N-(2-phenylethyl)-(9Cl), 5-alkyl substi 0616 mitogen-activated protein kinase (MAP) inhibitors tuted 2-arylaminophenylacetic acid and its derivatives (e.g., which include without limitation benzenesulfonamide, N-2- celecoxib (Celebrex(R), rofecoxib (Vioxx(R), etoricoxib (Ar 3-(4-chlorophenyl)-2-propenylmethylaminomethyl coxiaR), lumiracoxib (Prexige(R), Valdecoxib (Bextra(R) or phenyl-N-(2-hydroxyethyl)-4-methoxy-(9Cl). 5-alkyl-2-arylaminophenylacetic acid). 0617 MDM2 inhibitors which include without limitation 0601 cRAF kinase inhibitors which include without limi trans-4-iodo. 4'-boranyl-chalcone. tation 3-(3,5-dibromo-4-hydroxybenzylidene)-5-iodo-1,3- 0618 MEK inhibitors which include without limitation dihydroindol-2-one and benzamide, 3-(dimethylamino)-N- butanedinitrile, bisamino 2-aminophenyl)thiomethylene 3-(4-hydroxybenzoyl)amino-4-methylphenyl-(9Cl). (9Cl). 0602 cyclin dependent kinase inhibitors which include 0619 MMP inhibitors which include without limitation without limitation olomoucine and its derivatives, purvalanol Actinonin, epigallocatechingallate, collagen peptidomimetic B, roascovitine ((R), indirubin, kenpaullone, purv and non-peptidomimetic inhibitors, tetracycline derivatives alanol A and indirubin-3'-monooxime. marimastat (Marimastat(R), prinomastat, incyclinide (Met 0603 cysteine protease inhibitors which include without astat(R), shark cartilage extract AE-941 (Neovastat(R), Tano limitation 4-morpholinecarboxamide, N-(1S)-3-fluoro-2- mastat, TAA211, MMI270B or AAJ996. oXo-1-(2-phenylethyl)propylamino-2-oxo-1-(phenylm 0620 mTor inhibitors which include without limitation ethyl)ethyl-(9Cl). rapamycin (RapamuneR), and analogs and derivatives 0604 DNA intercalators which include without limitation thereof, AP23573 (also known as ridaforolimus, deforolimus, (Mithracinr) and daptomycin (Cubicin R). or MK-8669), CCI-779 (also known as ) 0605 DNA strand breakers which include without limita (Torisel R) and SDZ-RAD. tion bleomycin (Blenoxane(R). 0621 NGFR tyrosine kinase inhibitors which include 06.06 E3 ligase inhibitors which include without limita without limitation tyrphostin AG 879. tion N-((3,3,3-trifluoro-2-trifluoromethyl)propionyl)sulfa 0622 p38 MAP kinase inhibitors which include without nilamide limitation Phenol, 4-4-(4-fluorophenyl)-5-(4-pyridinyl)- 0607 EGF Pathway Inhibitors which include, without 1H-imidazol-2-yl)-(9Cl), and benzamide, 3-(dimethy limitation tyrphostin 46, EKB-569, erlotinib (Tarceva(R), lamino)-N-3-(4-hydroxylbenzoyl)amino-4-methylphe gefitinib (IressaR), lapatinib (Tykerb(R) and those com nyl-(9Cl). pounds that are generically and specifically disclosed in WO 0623 p56 tyrosine kinase inhibitors which include with 97/02266, EP 0564409, WO 99/03854, EP 0520 722, EP 0 out limitation damnacanthal and tyrphostin 46. 566 226, EP 0 787 722, EP 0 837 063, U.S. Pat. No. 5,747, 0624 PDGF pathway inhibitors which include without 498, WO 98/10767, WO 97/30034, WO 97/49688, WO limitation tyrphostin AG 1296, tyrphostin 9, 1,3-butadiene-1, 97/38983 and WO 96/3398O. 1,3-tricarbonitrile, 2-amino-4-(1H-indol-5-yl)-(9Cl), ima 0608 farnesyltransferase inhibitors which include with tinib (GleevecR) and gefitinib (IressaR) and those com out limitation A-hydroxyfarnesylphosphonic acid, butanoic pounds generically and specifically disclosed in European acid, 2-(2S)-2-(2S,3S)-2-(2R)-2-amino-3-mercaptopro Patent No.: O 564 409 and PCT Publication No.: WO pylamino-3-methylpentylloxy-1-oxo-3-phenylpropyl 99/O3854. amino-4-(methylsulfonyl)-1-methylethylester (2S)-(9Cl). 0625 phosphatidylinositol 3-kinase inhibitors which and manumycin A. include without limitation wortmannin, and quercetin dihy 0609 Flk-1 kinase inhibitors which include without limi drate. tation 2-propenamide, 2-cyano-3-4-hydroxy-3,5-bis(1-me 0626 phosphatase inhibitors which include without limi thylethyl)phenyl-N-(3-phenylpropyl)-(2E)-(9Cl). tation cantharidic acid, cantharidin, and L-leucinamide 0610 glycogen synthase kinase-3 (GSK3) inhibitors 0627 protein phosphatase inhibitors which include with which include without limitation indirubin-3'-monooxime. out limitation cantharidic acid, cantharidin, L-P-bromotetra 0611 histone deacetylase (HDAC) inhibitors which misole oxalate, 2(5H)-furanone, 4-hydroxy-5-(hydroxym include without limitation suberoylanilide hydroxamic acid ethyl)-3-(1-oxohexadecyl)-(5R)-(9Cl) and (SAHA), 4-(2-amino-phenylcarbamoyl)-benzyl-carbamic benzylphosphonic acid. acid pyridine-3-ylmethylester and its derivatives, butyric 0628 PKC inhibitors which include without limitation acid, pyroxamide, trichostatin A, oXamflatin, apicidin, dep 1-H-pyrollo-2,5-dione, 3-1-3-(dimethylamino)propyl sipeptide, depudecin, trapoxin and compounds disclosed in 1H-indol-3-yl)-4-(1H-indol-3-yl)-(9Cl), Bisindolylmaleim WO O2/22577. ide IX, Sphinogosine, staurosporine, and Hypericin. 0612 I-kappa B-alpha kinase inhibitors (IKK) which 0629 PKC delta kinase inhibitors which include without include without limitation 2-propenenitrile, 3-(4-meth limitation rottlerin. ylphenyl)sulfonyl-(2E)-(9Cl). 0630 polyamine synthesis inhibitors which include with 0613 imidazotetrazinones which include without limita out limitation DMFO. tion temozolomide (Methazolastone(R), Temodar R and its 0631 proteasome inhibitors which include, without limi derivatives (e.g., as disclosed generically and specifically in tation aclacinomycin A, gliotoxin and bortezomib (Vel U.S. Pat. No. 5,260.291) and . cade(R). 0614 insulin tyrosine kinase inhibitors which include 0632 PTP1B inhibitors which include without limitation without limitation hydroxyl-2-naphthalenylmethylphospho L-leucinamide nic acid. 0633 protein tyrosine kinase inhibitors which include, 0615 c-Jun-N-terminal kinase (JNK) inhibitors which without limitation tyrphostin Ag 216, tyrphostin Ag 1288, include without limitation pyrazoleanthrone and epigallocat tyrphostin Ag 1295, geldanamycin, genistein and 7H-pyrollo echin gallate. 2,3-dipyrimidine derivatives of formula I as generically and US 2011/O160 159 A1 Jun. 30, 2011 53 specifically described in PCT Publication No.: WO Oestrogenine(R), Oestromenin(R), Oestromon R, Palestrol(R), O3/O13541 and U.S. Publication No.: 2008/O139587: Stilbestrol(R), Stilbetin R, Stilboestroform(R), Stilboestrol(R), SynestrinR), Synthoestrin R, VagestrolR), testosterone (De latestry1(R), Testoderm(R, Testolin R, Testostroval(R), Test

(I) ostroval-PAR, Testro AQR), prednisone (Delta-DomeR, Deltasone(R), Liquid PredR), Lisacort(R), Meticorten(R), Ora sone(R), Prednicen-M(R), Sk-Prednisone(R), SterapredR). Flu oxymesterone (Android-F(R), Halodrin R, Halotestin R, Ora Testryl(R), Ultandren(R), dromostanolone propionate (Drolban R, Emdisterone(R), MasteridR, Masteril.R., Mas teron(R), Masterone(R), Metholone(R), Permastril.R.), testolac tone (TeslacR), megestrolacetate (Magestin R, Maygace R, Megace(R), Megeron(R), Megestat(R), Megestil(R), Megestin R, NiaR), Niagestin R, Ovaban R, Ovarid R, Volidan R.), methyl 0634 Publication No.: 2008/0139587 discloses the vari prednisolone (Depo-MedrolR), Medlone 21(R), MedrolR), ous Substituents, e.g., R. R. etc. Meprolone.R., Metrocort(R), MetypredR, Solu-MedrolR), 0635 SRC family tyrosine kinase inhibitors which Summicort(R), methyl-testosterone (Android R, TestredR, include without limitation PP1 and PP2. Virilon(R), prednisolone (Cortalone.R., Delta-CortefR), 0636 Syk tyrosine kinase inhibitors which include with Hydeltra(R), Hydeltrasol R, Meti-derm(R), Prelone(R), triamci out limitation piceatannol. nolone (Aristocort(R), chlorotrianisene (AniseneR), Chlorot 0637 Janus (JAK-2 and/or JAK-3) tyrosine kinase inhibi risin(R), Clorestrolo(R), Clorotrisin(R), Hormonisene(R), Khlor tors which include without limitation tyrphostin AG 490 and trianizen(R), Merbentul R., Metace(R), Rianil.R., Tace(R), Tace 2-naphthyl vinyl ketone. Fn(R), Trianisestrol(R), hydroxyprogesterone (Delalutin R, 0638 which include without limitation isotretin GestivaTM), aminoglutethimide (Cytadren(R), Elipten R, oin (Accutane(R), Amnesteem,R), Cistane(R), Claravis(R), Orimeten(R), estramustine (EmcytR), medroxyprogestero SotretR) and (Aberel(R), Aknoten(R), AvitaR), neacetate (Provera R, Depo-ProveraR), leuprolide (Lupron(R), Renova R, Retin-AR, Retin-A MICROR, VesanoidR). Viadur(R), flutamide (Eulexin R), toremifene (Fareston(R), 0639 RNA polymerase II elongation inhibitors which and goserelin (ZoladeX(R). include without limitation 5,6-dichloro-1-beta-D-ribofura nosylbenzimidazole. (0646. In certain embodiments, the CDP-topoisomerase 0640 serine/threonine kinase inhibitors which include inhibitor conjugate, particle or composition is administered without limitation 2-aminopurine. in combination with an anti-microbial (e.g., leptomycin B). 0641 sterol biosynthesis inhibitors which include without 0647. In another embodiment, the CDP-topoisomerase limitation squalene epoxidase and CYP2D6. inhibitor conjugate, particle or composition is administered 0642 VEGF pathway inhibitors which include without in combination with an agent or procedure to mitigate poten limitation anti-VEGF antibodies, e.g., bevacizumab, and tial side effects from the agent compositions such as cystisis, small molecules, e.g., Sunitinib (Sutent(R), Sorafinib (Nexa hypersensitivity, , and Vomiting. var(R), ZD6474 (also known as vandetanib) (ZactimaTM), 0648. Cystisis can be mitigated with an agent that SU6668, CP-547632, AV-951 (tivozanib) and AZD2171 (also increases urinary excretion and/or neutralizes one or more known as cediranib) (RecentinTM). urinary metabolite. For example, cystisis can be mitigated or 0643. Examples of chemotherapeutic agents are also treated with MESNA. described in the Scientific and patent literature, see, e.g., 0649. Diarrhea may be treated with antidiarrheal agents Bulinski (1997) J. Cell Sci. 110:3055-3064; Panda (1997) including, but not limited to opioids (e.g., codeine (Codic Proc. Natl. Acad. Sci. USA 94.10560-10564: Muhlradt ept(R), Coducept(R), oxicodeine, percocet, paregoric, tincture (1997) Cancer Res. 57:3344-3346; Nicolaou (1997) Nature of opium, diphenoxylate (Lomotil(R), diflenoXin), and lop 387:268-272; Vasquez (1997) Mol. Biol. Cell. 8:973-985; eramide (Imodium A-DR), bismuth subsalicylate, lanreotide, Panda (1996).J. Biol. Chem. 271:29807-29812. vapreotide (Sanvar(R), Sanvar IRR), motiln antagonists, 0644. In some embodiment, the CDP-topoisomerase COX2 inhibitors (e.g., celecoxib (Celebrex(R), glutamine inhibitor conjugate, particle or composition is administered (NutreStore?R), thalidomide (Synovir R, Thalomid(R), tradi instead of another topoisomerase inhibitor, e.g., instead of a tional antidiarrhea remedies (e.g., kaolin, pectin, berberine topoisomerase inhibitor as a first line therapy or a second line and muscarinic agents), octreotide and DPP-IV inhibitors. therapy. For example, the CDP-topoisomerase inhibitor con 0650 DPP-IV inhibitors employed in the present inven jugate, particle or composition can be used instead of any of tion are generically and specifically disclosed in PCT Publi the following topoisomerase inhibitors: a topoisomerase I cation Nos.: WO98/19998, DE 1961.6486A1, WO 00/34241 inhibitor, e.g., camptothecin, irinotecan, SN-38, topotecan, and WO95/15309. lamellarin D; a topoisomerase II inhibitor, e.g., etoposide, 0651 Nausea and vomiting may be treated with anti tenoposide, doxorubicin. emetic agents such as dexamethasone (Aeroseb-DeX(R), Alba 0645. In some cases, a hormone and/or steriod can be Dex R, Decaderm R), Decadrol R, Decadron R, Decasone(R), administered in combination with a CDP-topoisomerase Decaspray(R), Deenar(R), Deronil R, Dex-4R), Dexace(R), Dex inhibitor conjugate, particle or composition. Examples of amethR), DeZone(R), Gammacorten(R), HexadrolR), Maxi hormones and steroids include: 17a-ethinylestradiol (Esti dex(R, Sk-Dexamethasone(R), metoclopramide (ReglanR), nyl(R), Ethinoral R. Feminone(R), Orestralyn R.), diethylstil diphenylhydramine (Benadryl(R), SK-Diphenhydramine(R). bestrol (Acnestrol(R), Cyren AR, Deladumone(R), Diastyl(R), Iorazepam (Ativan(R), ondansetron (ZofranR), prochlorpera Domestrol R, Estrobene(R), EstrobeneR, Estrosyn R, Fona zine (Bayer A 173R), Buccastem.R., Capazine(R), CombidR, tol(R), Makarol(R), Milestrol(R), Milestrol(R), Neo-Oestronol IR), Compazine(R), ComproR), Emelent(R), Emetiral R, Eskatrol(R), US 2011/O160 159 A1 Jun. 30, 2011 54

Kronocin R, Meterazin R, Meterazin Maleate(R), Metera chemotherapeutic agents which are administered one every zine(R), Nipodal(R), Novamin R, Pasotomin R, Phenotil(R), Ste three weeks include: an antimetabolite (e.g., floXuridine metil(R), Stemzine(R, Tementil(R), Temetid R, Vertigon(R), thi (FUDF(R), pemetrexed (ALIMTAR), 5FU (Adrucil(R), Efu ethylperazine (Norzine(R), Torecan R), and dronabinol deX(R), FluoropleX(R)); an anthracycline (e.g., daunorubicin (MarinolR). (Cerubidine(R), Rubidomycin R), epirubicin (Ellence(R), ida 0652. In some embodiments, the CDP-topoisomerase rubicin (Idamycin R.), mitoxantrone (Novantrone(R), valrubi inhibitor conjugate, particle or composition is administered cin (Valstar(R)); a Vinca alkaloid (e.g., vinblastine (Velban R, in combination with an immunosuppressive agent. Immuno Velsar R), Vincristine (Vincasar R, OncovinR), vindesine (El Suppressive agents Suitable for the combination include, but disine(R) and vinorelbine (Navelbine(R)); a taxane (e.g., pacli are not limited to (TysabriR), (Imu taxel, docetaxel, larotaxel and cabazitaxel); and a platinum ran R.), mitoxantrone (Novantrone.R.), mycophenolate mofetil based agent (e.g., cisplatin (PlatinolR), carboplatin (Cellcept(R), cyclosporins (e.g., Cyclosporin A (Neoral(R), (Paraplatr), ParaplatinR), oxaliplatin (EloxatinR)). SandimmunR), Sandimmune(R), SangCya(R), cacineurin 0658. In another embodiment, the CDP-topoisomerase inhibitors (e.g., (Prograf R, Protopic(R), inhibitor conjugate, particle or composition is administered (Rapamune(R), everolimus (Afinitor(R), cyclophosphamide once every two weeks in combination with one or more addi (Clafen R, Cytoxan R, Neosar(R), or methotrexate (Abitrex tional chemotherapeutic agent that is administered orally. For ate(R). Folex(R), Methotrexate R, MexateR)), , example, the CDP-topoisomerase inhibitor conjugate, par mycophenolate mofetil (CellCept(R), ticle or composition can be administered once every two (Myfortic(R), anti-CD3 antibody, anti-CD25 antibody (e.g., weeks in combination with one or more of the following (Simulect(R) or (Zenapax(R)), and chemotherapeutic agents: capecitabine (Xeloda(R), estramus anti-TNFC. antibody (e.g., (Remicade(R) or adali tine (EmcytR), erlotinib (Tarceva(R), rapamycin (Rapam mumab (HumiraR)). une(R), SDZ-RAD, CP-547632: AZD2171, sunitinib 0653. In some embodiments, a CDP-topoisomerase (Sutent(R), Sorafenib (Nexavar R) and everolimus (Afini inhibitor conjugate, particle or composition is administered tor(R). in combination with a CYP3A4 inhibitor (e.g., ketoconazole 0659. The actual dosage of the CDP-topoisomerase (Nizoral(R), Xolegel(R), itraconazole (SporanoXR), clarithro inhibitor conjugate, particle or composition and/or any addi mycin (Biaxin R), atazanavir (ReyataZ(R), nefazodone (Ser tional chemotherapeutic agent employed may be varied Zone(R), Nefadar R), saquinavir (Invirase(R), tellithromycin depending upon the requirements of the Subject and the sever (Ketek(R), ritonavir (Norvir R), amprenavir (also known as ity of the condition being treated. Determination of the proper Agenerase, a prodrug version is foSamprenavir (LexivaR), dosage for a particular situation is within the skill of the art. TelzirR), indinavir (Crixivan(R), nelfinavir (Viracept(R), Generally, treatment is initiated with Smaller dosages which delavirdine (Rescriptor R) or Voriconazole (Vfend(R)). are less than the optimum dose of the compound. Thereafter, 0654 When employing the methods or compositions, the dosage is increased by Small amounts until the optimum other agents used in the modulation of tumor growth or effect under the circumstances is reached. in a clinical setting, Such as , can also 0660. In some embodiments, when a CDP-topoisomerase be administered as desired. inhibitor conjugate, particle or composition is administered 0655 When formulating the pharmaceutical composi in combination with one or more additional chemotherapeu tions featured in the invention the clinician may utilize pre tic agent, the additional chemotherapeutic agent (or agents) is ferred dosages as warranted by the condition of the subject administered at a standard dose. For example, a standard being treated. For example, in one embodiment, a CDP-to dosage for cisplatin is 75-120 mg/m administered every poisomerase inhibitor conjugate, particle or composition may three weeks; a standard dosage for carboplatin is within the be administered at a dosing schedule described herein, e.g., range of 200-600mg/moran AUC of 0.5-8 mg/mlxmin; e.g., once every one, two, three or four weeks. at an AUC of 4-6 mg/mlxmin; a standard dosage for irinote 0656 Also, in general, a CDP-topoisomerase inhibitor can is within 100-125 mg/m, once a week; a standard dosage conjugate, particle or composition and an additional chemo for gemcitabine is within the range of 80-1500 mg/m admin therapeutic agent(s) do not have to be administered in the istered weekly; a standard dose for UFT is within a range of same pharmaceutical composition, and may, because of dif 300-400 mg/m per day when combined with leucovorin ferent physical and chemical characteristics, have to be administration; a standard dosage for leucovorin is 10-600 administered by different routes. For example, the CDP-to mg/m administered weekly. poisomerase inhibitor conjugate, particle or composition may 0661 The disclosure also encompasses a method for the be administered intravenously while the chemotherapeutic synergistic wherein a CDP-topoi agent(s) may be administered orally. The determination of the Somerase inhibitor conjugate, particle or composition is mode of administration and the advisability of administra administered in combination with an additional chemothera tion, where possible, in the same pharmaceutical composi peutic agent or agents. tion, is well within the knowledge of the skilled clinician. The 0662. The particular choice of conjugate, particle or com initial administration can be made according to established position and anti-proliferative cytotoxic agent(s) or radiation protocols known in the art, and then, based upon the observed will depend upon the diagnosis of the attending physicians effects, the dosage, modes of administration and times of and their judgment of the condition of the subject and the administration can be modified by the skilled clinician. appropriate treatment protocol. 0657. In one embodiment, a CDP-topoisomerase inhibitor 0663 If the CDP-topoisomerase inhibitor conjugate, par conjugate, particle or composition is administered once every ticle or composition and the chemotherapeutic agent(s) and/ three weeks and an additional therapeutic agent (or additional or radiation are not administered simultaneously or essen therapeutic agents) may also be administered every three tially simultaneously, then the initial order of administration weeks for as long as treatment is required. Examples of other of the CDP-topoisomerase inhibitor conjugate, particle or US 2011/O160 159 A1 Jun. 30, 2011 composition, and the chemotherapeutic agent(s) and/or radia hood gastric (stomach) cancer; hepatocellular (liver) cancer tion, may be varied. Thus, for example, the CDP-topoi including childhood hepatocellular (liver) cancer; pancreatic Somerase inhibitor conjugate, particle or composition may be cancer including childhood pancreatic cancer, sarcoma, rhab administered first followed by the administration of the che domyosarcoma; pancreatic cancer, islet cell; rectal cancer, motherapeutic agent(s) and/or radiation; or the chemothera and Small intestine cancer, peutic agent(s) and/or radiation may be administered first 0671 Endocrine cancers such as islet cell carcinoma (en followed by the administration of the CDP-topoisomerase docrine pancreas); adrenocortical carcinoma including child inhibitor conjugate, particle or composition. This alternate hood adrenocortical carcinoma; gastrointestinal carcinoid administration may be repeated during a single treatment tumor, parathyroid cancer, pheochromocytoma; pituitary protocol. The determination of the order of administration, tumor, thyroid cancer including childhood thyroid cancer, and the number of repetitions of administration of each thera childhood multiple endocrine neoplasia syndrome; and child peutic agent during a treatment protocol, is well within the hood carcinoid tumor; knowledge of the skilled physician after evaluation of the 0672 Eye cancers such as intraocular melanoma; and ret disease being treated and the condition of the Subject. inoblastoma; 0664 Thus, in accordance with experience and knowl 0673 Musculoskeletal cancers such as Ewing's family of edge, the practicing physician can modify each protocol for tumors; osteosarcoma/malignant fibrous histiocytoma of the the administration of a component (CDP-topoisomerase bone; rhabdomyosarcoma including childhood rhabdomyo inhibitor conjugate, particle or composition, anti-neoplastic sarcoma; Soft tissue sarcoma including childhood soft tissue agent(s), or radiation) of the treatment according to the indi sarcoma; clear cell sarcoma of tendon sheaths; and uterine vidual subject's needs, as the treatment proceeds. SarCOma, 0665. The attending clinician, in judging whether treat 0674 Breast cancer Such as breast cancer and pregnancy ment is effective at the dosage administered, will consider the including childhood and male breast cancer, general well-being of the subject as well as more definite 0675 Neurologic cancers such as childhood brain stem signs such as relief of disease-related symptoms, inhibition of glioma; brain tumor; childhood cerebellar astrocytoma; tumor growth, actual shrinkage of the tumor, or inhibition of childhood cerebral astrocytoma/malignant glioma; child metastasis. Size of the tumor can be measured by standard hood ependymoma; childhood medulloblastoma; childhood methods such as radiological studies, e.g., CAT or MRI scan, pineal and Supratentorial primitive neuroectodermal tumors; and Successive measurements can be used to judge whether or childhood visual pathway and hypothalamic glioma; other not growth of the tumor has been retarded or even reversed. childhood brain cancers; adrenocortical carcinoma; central Relief of disease-related symptoms such as pain, and nervous system lymphoma, primary; childhood cerebellar improvement in overall condition can also be used to help astrocytoma; neuroblastoma; craniopharyngioma; spinal judge effectiveness of treatment. cord tumors; central nervous system atypical teratoid/rhab 0.666 Indications doid tumor, central nervous system embryonal tumors; and 0667 The disclosed CDP-topoisomerase inhibitor conju Supratentorial primitive neuroectodermal tumors including gates, particles and compositions are useful in treating pro childhood and pituitary tumor; liferative disorders, e.g., treating a tumor, e.g., a primary 0676 Genitourinary cancers such as bladder cancer tumor, and/or metastases thereof, wherein the tumor is a including childhood bladder cancer, renal cell (kidney) can primary tumor or a metastases thereof, e.g., a cancer cer; ovarian cancer including childhood ovarian cancer, ova described herein or a metastases of a cancer described herein. rian epithelial cancer, ovarian low malignant potential tumor; 0668. The methods described herein can be used to treat a penile cancer, prostate cancer, renal cell cancer including Solid tumor, a soft tissue tumor or a liquid tumor. Exemplary childhood renal cell cancer; renal pelvis and ureter, transi Solid tumors include malignancies (e.g., Sarcomas and carci tional cell cancer, testicular cancer, urethral cancer, vaginal nomas (e.g., adenocarcinoma or Squamous cell carcinoma)) cancer, Vulvar cancer; cervical cancer, Wilms tumor and of the various organ systems, such as those of brain, lung, other childhood kidney tumors; endometrial cancer, and ges breast, lymphoid, gastrointestinal (e.g., colon), and genitouri tational trophoblastic tumor; nary (e.g., renal, urothelial, or testicular tumors) tracts, phar 0677 Germ cell cancers such as childhood extracranial ynx, prostate, and ovary. Exemplary adenocarcinomas germ cell tumor, extragonadal germ cell tumor, ovarian germ include colorectal cancers, renal-cell carcinoma, liver cancer, cell tumor; and testicular cancer, non-Small cell carcinoma of the lung, and cancer of the Small 0678 Head and neck cancers such as lip and oral cavity intestine. The disclosed methods are also useful in evaluating cancer, childhood oral cancer; hypopharyngeal cancer, laryn or treating Soft tissue tumors such as those of the tendons, geal cancer including childhood laryngeal cancer, metastatic muscles or fat, and liquid tumors. squamous neck cancer with occult primary; mouth cancer, 0669. The methods described herein can be used with any nasal cavity and paranasal sinus cancer, nasopharyngeal can cancer, for example those described by the National Cancer cer including childhood nasopharyngeal cancer, oropharyn Institute. The cancer can be a carcinoma, a sarcoma, a geal cancer, parathyroid cancer, pharyngeal cancer; salivary myeloma, a leukemia, a lymphoma or a mixed type. Exem gland cancer including childhood salivary gland cancer, plary cancers described by the National Cancer Institute throat cancer, and thyroid cancer, include: 0679 Hematologic/blood cell cancers such as a leukemia 0670 Digestive/gastrointestinal cancers such as anal can (e.g., acute lymphoblastic leukemia in adults and children; cer; bile duct cancer; extrahepatic bile duct cancer; appendix acute myeloid leukemia, e.g., in adults and children; chronic cancer, carcinoid tumor, gastrointestinal cancer, colon can lymphocytic leukemia; chronic myelogenous leukemia; and cer; colorectal cancer including childhood colorectal cancer, hairy cell leukemia); a lymphoma (e.g., AIDS-related lym esophageal cancer including childhood esophageal cancer, phoma; cutaneous T-cell lymphoma; Hodgkin’s lymphoma gallbladder cancer, gastric (stomach) cancer including child including Hodgkin's lymphoma in adults and children; US 2011/O160 159 A1 Jun. 30, 2011 56

Hodgkin's lymphoma during pregnancy; non-Hodgkin’s tumors refractory to standard therapy, or for which no stan lymphoma including non-Hodgkin’s lymphoma in adults and dard curative or palliative therapy existed, were eligible for children; non-Hodgkin's lymphoma during pregnancy; this trial. Prior treatment with topoisomerase inhibitors was mycosis fungoides; Sezary syndrome; Waldenstrom's mac allowed. Main eligibility criteria included male or female roglobulinemia; and primary central nervous system lym patients, at least 18 years of age, with advanced, histologi phoma); and other hematologic cancers (e.g., chronic myelo cally confirmed Solid tumor refractory to standard treatment, proliferative disorders; multiple myeloma/plasma cell or for which no standard therapy existed, measurable or ; myelodysplastic syndromes; and myelodysplas evaluable disease, Eastern Cooperative Group tic/myeloproliferative disorders); (ECOG) Performance Status s2, acceptable organ and bone 0680 Lung cancer Such as non-Small cell lung cancer, and marrow function, no evidence of clinically significant con Small cell lung cancer, duction abnormalities or ischemia, and ejection fraction 0681 Respiratory cancers such as malignant mesothe 245%. Prior , radiation therapy, or investiga lioma including malignant mesothelioma in adults and chil tional therapy had to be completed within a prescribed inter dren; malignant thymoma; childhood thymoma; thymic car val before enrollment, could not have included high dose cinoma; bronchial adenomas/carcinoids including childhood chemotherapy with autologous stem cell rescue bone marrow bronchial adenomas/carcinoids; pleuropulmonary blastoma; transplantation, and patients could not have been refractory to non-Small cell lung cancer, and Small cell lung cancer, prior treatment with a topoisomerase I inhibitor. 0682 Skin cancers such as Kaposi's sarcoma; Merkel cell (0692. This trial was conducted at City of Hope (COH) carcinoma; melanoma; and childhood skin cancer, (Duarte, Calif.) following approval of the Clinical Protocol 0683 AIDS-related malignancies: Review and Monitoring Committee (CPRMC) and the Insti 0684 Other childhood cancers, unusual cancers of child tutional Review Board (IRB). After the initial 18 patients hood and cancers of unknown primary site; were enrolled, an additional site was added at TGen (Scotts 0685 and metastases of the aforementioned cancers can dale, Ariz.) following the approval of WIRB (Western Insti also be treated or prevented in accordance with the methods tutional Review Board). described herein. 0693 Study design and drug administration. This was an 0686. The CDP-topoisomerase inhibitor conjugates, par open-labeled, single-arm dose-escalation phase 1 study of ticles and compositions described herein are particularly CRLX101. In part 1, CRLX101 was administered at 6, 12, or Suited to treat accelerated or metastatic cancers of gastric 18 mg/m on a weekly schedule three weeks per month. In cancer, colorectal cancer, non-Small cell lung cancer, ovarian part 2, CRLX101 was administered at 12, 15 or 18 mg/m on cancer, and breast cancer. an every other week schedule. 0687 In one embodiment, a method is provided for a 0694. In part 1, CRLX101 was administered as an intra combination treatment of a cancer, Such as by treatment with venous infusion over 90 minutes on days 1, 8, and 15 followed a CDP-topoisomerase inhibitor conjugate, particle or compo by a 7 day rest period (28 day cycle). In part 2, CRLX101 was sition and a second therapeutic agent. Various combinations administered as an intravenous infusion over 90 minutes on are described herein. The combination can reduce the devel days 1 and 15, every 28 days. CRLX101 was administered as opment of tumors, reduces tumor burden, or produce tumor an intravenous infusion over 60 minutes on days 1 and 15. regression in a mammalian host. every 28 days for the last six patients in part 2 of the Phase 1 0688. Unless otherwise defined, all technical and scien study. tific terms used herein have the same meaning as commonly (0695) The starting dose in part 1 was 6 mg/m. Dose understood by one of ordinary skill in the art to which this escalation was done using an accelerated Simon design (Si invention belongs. All publications, patent applications, pat monet al., “Accelerated Titration Designs for Phase I Clinical ents, and other references mentioned herein are incorporated Trials in Oncology”J Natl Cancer Inst. 89:1138-47, 1997) by reference in their entirety. In case of conflict, the present with a modified Fibonacci dose escalation scheme. In brief, specification, including definitions, will control. In addition, patients were accrued in cohorts of one at escalating doses the materials, methods, and examples are illustrative only and until the occurrence of a dose limiting toxicity (DLT) in the not intended to be limiting. first cycle. Once a DLT occurred, additional patients were accrued in that dosage group to provide for either 3 or 6 EXAMPLES patients as provided in a standard dose escalation with the Example 1 intent of determining the maximum tolerated dose. Concur rent accrual was allowed within the same dose level. No Human Phase 1 Study of CRLX101 intrapatient dose escalation was permitted. 0689. The below example describes the first human phase 0696 Toxicity Assessment. Toxicity was graded accord 1 study of CRLX101. The study was composed of two parts. ing to the National Cancer Institute (NCI) common toxicity Part 1 of the Phase 1 study had a primary objective to deter criteria (CTCAE) version 3.0. The DLT in a given patient was mine the safety profile, toxicity, and pharmacokinetics of defined as any treatment-related grade III non-hematologic CRLX101 when administered weekly for 3 consecutive toxicity, any grade IV hematologic toxicity, or persisting tox weeks of every 4-week cycle (the initial dosing regimen, icities of any grade requiring delay of scheduled treatment by sometimes referred to herein as “weekly x3’). In part 2, after more than 2 weeks. DLT was based on the first course of the first twelve patients were enrolled, an every other week treatment. schedule was initiated for the remainder of the Phase 1 pro 0697 Rules for dose escalation. One patient was treated at gram (sometimes referred to herein as “biweekly’). each dose level. If a DLT attributable to the study drug(s) was 0690 Patients and Methods experienced, up to 5 additional patients were Subsequently 0691 Eligibility Criteria. Patients with histologically or treated at that dose level. If no additional DLTs were observed cytologically confirmed metastatic or unresectable Solid at the expanded dose level (i.e., at most /6 with an attributable US 2011/O160 159 A1 Jun. 30, 2011 57

DLT), the dose was escalated. Escalation was terminated hematologic toxicities that did not recover to grade 1 before when two of six patients experienced any DLT attributable to the next treatment, then the dose was reduced by 50% in the study drug at a given dose level. The maximally tolerated Subsequent cycles. If there were conflicting dose attenuations dose (MTD) was defined as the dose level preceding the dose by hematologic and non-hematologic toxicity, the greater at which 22/6 patients experienced a first-course DLT. Treat dose reduction was applied. All dose modifications were per ment was continued in an individual patient for a total of 6 manent. Criteria for removal from treatment included disease cycles at the same dose level if no DLT was observed and if progression, clinical progression, excessive toxicity, or clinical benefit was observed. Therapy was discontinued in patient withdrawal. any patient if excessive toxicity was experienced. No intra 0702 Plasma/urine sampling and analysis. Whole blood patient dose escalation was permitted. Patients who com samples (5 mL) were collected in heparinized tubes at the pleted 6 cycles with clinical benefit had the option of continu following times during cycle one: pre-dose, during dosing at ing treatment at the same dose level every other week. 30 minutes, 60 minutes and just prior to end of administration 0698 Safety and efficacy evaluations. Patients were seen, (90 minutes), and then at 0.25, 0.5, 1, 2, 3, 4, 6, 8, 24, (48-72 examined and a complete blood count with differential and hr optional), 168 hours (1 week), and 336 hours (only for serum chemistry was obtained before each dose. Radio patients on biweekly schedule) following the first dose. A graphic assessments of tumor response (as evaluated by the spot trough PK blood sample was collected prior to dosing on Response Evaluation Criteria in Solid Tumor criteria Day 1 and Day 15 of every cycle. Plasma was separated by RECIST) were performed every two cycles (Therasse et al., centrifugation at 1,300xg for 10 minutes at 4°C. Plasma was “New Guidelines to Evaluate the Response to Treatment in immediately frozen at -20°C., and stored frozen until analy Solid Tumors. European Organization for Research and sis. For determination of urinary excretion of polymer con Treatment of Cancer, National Cancer Institute of the United jugated and unbound CPT, total urine collections were per States, National Cancer Institute of Canada' J. Natl. Cancer formed following dose 1 of the first cycle (0-24 and 24-48 Inst. 92:205-16, 2000). hours post administration. A spot urine sample (15 mL) was 0699 Treatment modification. Patients who experienced a collected at 8, 24, 48, 168 (1 wk), and 336 hours (pre-second DLT during the initial cycle, or a severe or life threatening dose) for more accurate determination of total to free CPT non-hematologic toxicity at any time during the study, were ratio. Once each urine collection was complete, the actual dose reduced at the next dose administration when their DLT collection times and total Volume of urine was recorded and a returned to Grade 1. If a non-hematologic DLT occurred in 5-10 mL aliquot of each was frozen for subsequent drug level the interval between dose administrations within a cycle, and analysis. the toxicity was not life threatening and resolved rapidly, the 0703 Samples were extracted and analyzed by LC-MS/ next dose administration within the cycle was to be at the next MS (liquid chromatography-mass spectrometry/mass spec lower dose level. If a hematologic DLT occurred in the inter trometry) using a validated method. In order to determine Val between dose administrations withina cycle, the next dose total CPT in samples, a 20 uL, aliquot was incubated with 5 administration was to be at the next lower dose level, even if uL, 0.2N NaOH for 1 hour to release all CPT from the poly the blood cell counts recovered by the date of the next sched mer conjugate. The solution was acidified with 7 uI, 44% uled administration. Dosing was to be held for any non formic acid. Proteins were precipitated by addition of 160 uL hematologic toxicity grade 22, except for grade 2 fatigue and cold (<-20°C.) methanol containing the internal standard anorexia. After the first cycle, dose modifications were based 9-nitro camptothecin (9-NC) at a concentration of 8 ng/mL, on interval toxicity, and and granulocyte counts were incubation for 30 minutes on ice, and centrifugation at 14,000 obtained on the day of treatment. rpm. The resulting Supernatant was diluted with an equal 0700 Hematologic toxicity: On day 1 of a cycle, admin volume of 0.5 mM ammonium acetate buffer, pH 3.5, and istration of CRLX101 required an absolute granulocyte count analyzed by LC-MS/MS. (AGC) of 21500 and platelet count of 2100,000. On day 8, 0704. In order to determine unconjugated CPT in samples, if the AGC was 21000 and platelet count 275,000, then a 20 L, aliquot was acidified with 3 ul. 44% formic acid. CRLX101 was given at full dose and in the same manner for Proteins were precipitated by addition of 160 uL cold (<-20° day 15. If the AGC was <1000, the treating physicians could C.) methanol containing 9-NC at a concentration of 8 ng/mL, delay the dose of CRLX101 on days 8 and 15. If unacceptable incubation for 30 minutes on ice, and centrifugation at 14,000 hematologic toxicity persisted >7 days, the CRLX101 dose rpm. The resulting Supernatant was diluted with an equal was reduced to the next lower dose level. Similar standards volume of 0.5 mM ammonium acetate buffer, pH 3.5 and were used in the biweekly part. analyzed by LC-MS/MS. 0701. Non-hematologic toxicity: Any grade 2 toxicity that (0705 LC-MS/MS was performed on an Agilent 1100 was intolerable to the patients, or any grade 3 or greater series HPLC system (Palo Alto, Calif.) coupled to a Micro non-hematologic toxicity that was attributed to CRLX101, mass Quattro Ultima Triple Quadrupole Mass Spectrometer had to return to grade 1 before a new cycle of treatment was (Micromass, Inc., Beverly, Mass.). HPLC separation was started. Any treatment delay >2 weeks because of toxicity due achieved using a Synergy Hydro-RP 4 um 75x2 mm analyti to CRLX101 would result in the patient being removed from cal column (Phenomenex, Torrance, Calif.) preceded by a Cs the study. If a patient experienced any grade 3 genitourinary guard column (Phenomenex, Torrance, Calif.). The isocratic (GU) toxicity, the dose of CRLX101 was reduced to the next mobile phase consisted of 34% acetonitrile, 66% 0.5 mM lower dose level, as CPT has been reported to cause hema ammonium acetate, pH 3.5 at a flow rate of 0.2 mL/min at turia, cystitis or other GU toxicities, which can be irrevers room temperature. MassLynx version 3.5 software was used ible. If the patient experienced grade 3 non-hematologic tox for data acquisition and processing. icities that did not recover to grade 1 before the next 0706. Immunohistochemical analysis of topoisomerase I treatment, treatment was held until recovery and then reduced expression. Ascites fluid from one patient with ovarian cancer by one dose level. If the patient experienced grade 4 non was obtained pre-treatment and on day 2 post treatment with US 2011/O160 159 A1 Jun. 30, 2011

CRLX101 at 6 mg/m. Cells were pelleted by centrifugation are summarized in Table 1. The patients had a variety of solid and the pellet frozen. The pellet was formalin fixed and par tumors, and lung cancer was the most common tumor type. affin embedded. Immunohistochemical staining was per All twenty-four patients were considered evaluable for tox formed on 5 um thick sections. Sections were deparaffinized in xylene followed by 100% ethanol. Samples were then icity having received two completed cycles of therapy. quenched in 3% hydrogen peroxide and pretreated to promote antigen retrieval by steam in DIVA/citrate buffer (pH 6.0, TABLE 1 Biocare Medical, Concord, Calif.) solution. After antigen retrieval, slides were incubated in Protein Block for 20 min Patient demographics utes. Slides were then incubated with primary antibody over Total number of patients 24 night at 4°C. Topoisomerase I antibody was rabbit polyclonal Number of evaluable patients 24 (100%) from Abcam (Cambridge, Mass.). Median age (range) 61 (46-79) (0707. The next day, slides were washed in Dako Buffer Gender-Malef Female 13.11 (DB) and incubated with the appropriate secondary antibod Ethnicity ies for 30 minutes at room temperature. After washes in DB, Caucasian 16 (66%) slides were incubated with the chromogen diaminobenzidine Asian 6 (25%) tetrahydrochloride (DAB), counterstained with hematoxylin, Others 2 (9%) and mounted. Performance status (ECOG) 0708 Topoisomerase I enzymatic activity assay. Lysates O 11 (46%) containing total cellular protein were made from the plain 12 (50%) frozen ascites cells according to the method of Minagawa et 2 1 (4%) al. (“Enhanced Topoisomerase I Activity and Increased Tumor types Topoisomerase II Alpha Content in Cisplatin-Resistant Can Lung 6 cer Cell Lines' Jpn. J Cancer Res. 88:1218–23, 1997), a Breast 3 procedure used specifically for frozen cells. Urinary/Renal 3 0709. The catalytic activity of topoisomerase I was deter Liver 2 mined by measuring the relaxation of Supercoiled plasmid Pancreatic 5 substrate DNA using the Topo I assay kit (TopoCEN, Port Ovarian 1 Thyroid 1 Orange, Fla.) following the manufacturer's instructions. Head and Neck 1 Briefly, reaction mixtures consisted of supercoiled plasmid GI 1 substrate DNA (0.5ug), whole cell lysate (0.25ug or 0.5ug, Endometrial 1 as indicated in data) and the assay buffer (final concentra Prior chemotherapy regimens tions: 10 mM Tris HCl pH 7.9, 1 mM EDTA, 150 mMNaCl, O 1 0.1% BSA, 0.1 mM spermidine, and 5% glycerol). Reaction 5 mixtures were incubated at 37° C. for 30 minutes, and termi 2 2 nated by adding 5 uL stop buffer/gel loading buffer. Samples 3 4 were loaded onto a 1% agarose gel in 1xTAE buffer (40 mM greater than or equal to 4 12 Tris base pH 8.3, adjusted in the 50x stock buffer using glacial acetic acid, 2.5 mM NaOAc, and 0.05 mM EDTA) and electrophoresed at 4-5 volts percentimeter for 3-4 hours. 0713 Tumor response. Patient dosing and data are shown Supercoiled plasmid DNA (0.5ug) and relaxed DNA (0.5ug) in Table 2. At the first dose schedule (part 1), one patient with provided by the Topo I assay kit were used as the control metastatic pancreatic cancer that had spread to lungs and liver markers. The gel was stained with 0.2 Lug/mL ethidium bro experienced stable disease and received compassionate treat mide for 20 minutes at room temperature, destained in water ment for a total of 22 cycles before disease progression. The for 20 minutes and photographed under ultraviolet (UV) light. results of a CT scan performed on this patient are shown in 0710) Pharmacokinetic and statistical analysis. Plasma FIGS. 1A and 1B. Four patients experienced prolonged (>6 concentration versus time data were analyzed using the months) SD (stable disease), one patient with renal cancer ADAPT II (Biomedical Simulations Resource, Los Angeles) with lung metastases, two patients with non-Small cell lung non-compartmental model. Results are Summarized using cancer patient, and one patient with adenocarcinoma of the descriptive statistics. pancreas. (Table 3). All patients had been heavily pretreated 0711 Results for metastatic disease (see Table 3). Although the protocol 0712 Patient Enrollment. Twenty-four patients were allowed for prior treatment with CPT, none of these patients enrolled from June 2006 to April 2010. Patient characteristics had received prior CPT treatment.

TABLE 2 Analysis of population and patient disposition. Number Number of Best Responses Number of Number of patients patients No. Cycles PtS. w. DR During therapy Pts requiring of completed completed Median AE leading to Discontinuation (all eligible dose modification patients. cycle 1 cycle 6 (range) discontinuation AE description patients) for toxicity 6 mg/m. 6 6 1 (17%) 2.0 (1-6) O 1 SD (17%) 0 weekly x 3 12 mg/m 3 3 1 (33%) 2.0 (2-6) 2 (67%) , cystitis 2 SD (67%) 1 (33%) weekly x 3 US 2011/O160 159 A1 Jun. 30, 2011 59

TABLE 2-continued Analysis of population and patient disposition. Number Number of Best Responses Number of Number of patients patients No. Cycles Pts. w. DR During therapy Pts requiring of completed completed Median AE leading to Discontinuation (all eligible dose modification patients. cycle 1 cycle 6 (range) discontinuation AE description patients) for toxicity 18 mg/m. 3 3 1 (33%) 2.0 (1-6) 2 (67%) Neutropenia, 2 SD (67%) 2 (67%) weekly x 3 thrombocytopenia, upper resp. tract infection 12 mg/m 3 3 O 3.0 (2-4) O O biweekly 15 mg/m. 6 6 1 (16%) 3.0 (2-6) 1 Cystitis 4 SD (67%) 1 (17%) biweekly 18 mg/m 3 3 O 2 (1-5) 1 Thrombocytopenia 1 SD (33%) 2 (67%) biweekly

Note: One patient still active - presently in cycle 5

0714 Four patients who completed 6 cycles of therapy 0718. One lung cancer patient experienced elevated amy continued to receive CRLX101 every other week on a com lase in laboratory tests but no clinical manifestation of pan passionate use basis. Abbreviations: AE=adverse event, creatitis. SD-stable disease, PD progressive disease, DR-drug 0719. One head and neck cancer patient (nasopharyngeal related, Pt patient with heavy previous chemotherapy) experienced delayed onset grade 2 transaminitis in cycle 2, day 1. TABLE 3 0720. One lung cancer patient experienced grade 2 hema tologic toxicity which resolved in two weeks. CRLX101 showed activity in this heavily pre-treated patient population with 4 long-term progression free 0721. At the highest dose tested, two patients experienced survivors, two of which showed minor responses. DLT hematological toxicities. The first patient, a breast can cer patient, who had metastatic disease to lung and other Duration organs and had previously received multiple chemotherapy Dosef of Prior regimens, developed grade 3 anemia, neutropenia, and Tumor Type Schedule Activity Prior Agents Response thrombocytopenia. This patient required platelet transfu Pancreatic 6 mg/m’ 22.8 mo 5-FU, cisplatin, 10 mo sions. The second patient had metastatic lung cancer, and had weekly x 3 PFS, CT gemcitabine, previously received carboplatin and paclitaxel with a brief scan 16% radiation Non-small 18 mg/m. 9.7 mo Getfitinib, 6 mo response. This patient developed grade 3 neutropenia, requir cell lung weekly x 3 PFS, CT carboplatin, ing dose reduction and granulocyte colony-stimulating factor scan 6% paclitaxel, (G-CSF) support. This hematological toxicity in 2 of 3 pemetrexed, vinorelbine, patients established 18 mg/m as the DLT level. gemictabine 0722. One pancreatic cancer patient experienced grade 2 Renal 12 mg/m/ 7.7 mo Sunitinib 18 mo anemia on cycle 5, day 1. weekly x 3 PFS, SD 0723. One patient with hepatocellular cancer passed away Non-small 15 mg/m. 12.0 mo Carboplatin, 7 mo cell lung biweekly PFS, SD paclitaxel, one week after cycle 3, day 1 due to progressive disease. experimental RX, 0724. One patient with lung cancer who had received mul pemetrexed, tiple prior regimens, developed grade 2 neutropenia on cycle gemcitabine, 3, day 15. This patient required a dose reduction and G-CSF vinorelbine, erlotinib Support. 0725 Based on the long terminal half-life of polymer con Abbreviations: PFS = progression free survival, SD = stable disease jugated, and especially unconjugated, CPT (see FIGS. 2A and 2B), the protocol was amended in part 2 to evaluate an every 0715 Toxicity Evaluation. All patients in the first twelve other week schedule. A total of 12 patients were treated on enrolled (weekly x3) experienced grade 3 or 4 toxicities at this schedule, three at 12 mg/m and six at 15 mg/m and three doses above 6 mg/m. The weekly x3 schedule was stopped at 18 mg/m. Except for one occurrence of grade 3 neutrope after these first twelve patients due to the severe toxicity nia, no grade 3/4 hematologic events were recorded at dose observed in all patients treated at doses higher than 6 mg/m. level 12 mg/m and 15 mg/m. At dose level 18 mg/m one One patient tolerated 9 mg/m on a weekly x3 schedule for grade 4 leukopenia, two grade 4 neutropenia, two grade 4 five cycles (three doses per cycle). thrombocytopenia and one grade 3 anemia were reported 0716 No patients in the second twelve patients enrolled at under hematological toxicities. This determined the DLT of the bi-weekly schedule experienced severe toxicity until the bone marrow suppression at 18 mg/m on this schedule. The MTD was reached. Grade 2 DLTs included the following. only other notable non hematologic event was a grade 3 0717. One bladder cancer patient experienced grade 2 ane hypersensitivity reaction. (Table 4). mia in cycle 2, day 8. This grade 2 hematologic toxicity 0726 Table 4 shows a summary of all grade 3/4 treatment resolved in two weeks. related toxicities for all evaluable patients. Four patients on US 2011/O160 159 A1 Jun. 30, 2011 60 the weekly schedule developed delayed onset (after cycle 4) CRLX101 is initially retained in plasma and avoids rapid first mild hematuria and mild dysuria that may have been related pass clearance. C, and AUCo., were linear across doses to treatment as previously reported for CPT (Muggia et al., and similar when normalized for dose/m. FIGS. 2A and 2B “Phase I of Weekly and Daily Treatment with Camptothecin (NSC-100880): Correlation with Preclinical Summarize pharmacokinetic parameters measured for poly Studies' Cancer Chemother Rep. 56:515-521, 1972). How mer conjugated and unconjugated CPT for both schedules. ever, upon evaluation by a urologist, cystitis could not be Unconjugated CPT was slowly released from CRLX101 as confirmed. Thus, at 18 mg/m biweekly two patients were reported shown by increasing plasma concentrations that peaked at who developed DLT, and the MTD for the biweekly schedulie was established as 20.2+9.7 hrs. Plasma concentrations of unconjugated CPT 15 mg/m. were significantly below the plasma concentrations of conju gated CPT at all timepoints, with unconjugated CPT account TABLE 4 ing for an average of 8.7-2.7% of total CPT plasma exposure. Treatment related grade 3.4 hematologic and non-hematologic adverse FIG. 2A shows average plasma time-concentration curves for events by dose cohort observed during all courses of therapy. the biweekly 12 mg/m cohort Systemic plasma clearance of conjugated CPT was 0.12+0.2 L/h, significantly below the DOSE Grade 3 Grade 4 kidney and liver blood flows in humans, and was also inde 6 mg/m’ pendent of dose. FIG. 2B shows average urinary excretion of Elevated CPK 1 O polymer conjugated and unconjugated CPT in the first 48 Fatigue 1 hours following CRLX101 administration. Urinary loss of yearsnia 1 total CPT was variable with an average of 22.8+12.1% of mg/m. dose excreted during the first 48 hours, of which 78+9% was hyponatremia 2 O in the conjugated form. Interestingly, urinary excretion of the P.gaits 2 O polymer conjugate was primarily in the first 24 hours (16. 4%+10.0% of dose) compared to the second 24 hours (1.5+1. Elytopenia i 3% of dose) post administration. Urinary excretion of uncon RN(KO)68 4 3 jugated CPT remained approximately constant over both 24 anemia 3 O hour periods (2.0+1.1% vs. 2.9-1.4% of dose). Toxicokinetic E. ti g analysis of the two schedules showed that the predicted rashenydration 1 O monthly exposure for conjugated and unconjugated CPT was SOB 1 O similar for 6 mg/m weekly vs. 12 mg/m bi-weekly and 12 sensitivity 1 O mg/m weekly vs. 15 mg/m bi-weekly; however, fewer patients experienced cycle one drug related adverse events with the bi-weekly regimen.

TABLE 5 Pharmacokinetic parameters and toxicokinetic summary. T12, B AUC Per cycle AUC # Pt with Dosef Cuang L hir mg/L/hr. mg/L/hr DRAE 2

Schedule N Bound Free Bound Free Bound Free Bound Free Grade. 3 6 mg/m. 6 3.55 + 0.10 + 31.1 + 43.7 + 114.4+ 11.9 + 343.2 + 35.7 + 2 (33%) weekly x 3 O46 0.06 5.2 146 21.5 7.0 64.5 21.1 12 mg/m 3 5.55 + 0.18 + 33.8 + 61.5 + 188.5 + 18.3 + 565.6+ 54.9 + 1 (33%) weekly x 3 1.33 O.O1 S.9 37.6 56.7 3.8 1701 11.4 18 mg/m 3 7.90 + 0.24 + 37.7 + 38.3 + 248.3 + 23.7+ 744.8 + 71.1 + 3 (100%) weekly x 3 1.18 OO6 6.2 4.9 29.2 5.9 87.5 17.8 12 mg/m 3 5.56+ 0.22 + 27.8 + 32.5 + 182.0 + 14.6+ 364.0 + 29.0 + () biweekly O.37 0.09 43 4.9 21.6 2.5 43.1 S.O 15 mg/m2 3 8.63 + 0.27+ 30.4 + 48.3 + 276.7 + 23.5 + 553.8 + 47.0 + 1 (33%) biweekly O.76 O.13 1.2 6.5 14.2 9.0 28.3 18.0 Values are in geometric means + standard deviation,

0727 Pharmacokinetic and toxicokinetic analysis. 0728 Correlative Studies. Ascites cells were collected Samples for pharmacokinetic analysis were collected from all from one patient with ovarian cancer pre-treatment and on patients during cycle 1. Results from this analysis are Sum Days 2 and 25 post-treatment. Pellets of these cells were frozen for later analysis. Levels of polymer conjugated and marized in FIGS. 2A, 2B and Table 5. The mean elimination unconjugated CPT were also determined in the ascites fluid half-lives were 31.8+5.7 hr and 43.8+9.7 hr for conjugated pre-treatment and on day 2 post-treatment. On day 2, the and unconjugated CPT respectively. Volume of distribution concentrations detected were 46.6 ug/L for conjugated CPT of the polymer conjugate was 4.2t1.1 liter and was indepen and 19.6 g/L for fully active, unconjugated CPT. For each dent of dose. The low volume of distribution suggests that amount of lysate used, there was less topoisomerase I US 2011/O160 159 A1 Jun. 30, 2011

unwinding activity (i.e., more remaining Supercoiled DNA) demonstrated Stable disease on CT scan evaluation at the end in the Day 2 samples than in the pretreatment or Day 25 of cycle 2. One pancreatic cancer patient remained stable for samples. This suggests an inhibitory effect of CRLX101 on 22.8 months. Serum and urine PK data from all the treated these cells at this early time point after administration. The patients indicated that the mean elimination half-lives for ascites cells were also used for immunohistochemistry (IHC) to assess the levels of topoisomerase I. Basic agreement was conjugated and unconjugated CPT were 31.8 hr and 43.8 hr, observed between the topoisomerase I activity assay and respectively. C, and AUCo., were linear across doses and topoisomerase IIHC. As seen in FIGS. 3A and 3B, there was similar when normalized for dose/m. The biweekly schedule a reduction of approximately 30% in staining in the nucleus of allowed for similar per cycle plasma exposure to be achieved ovarian cancer cells isolated from the patient's ascites fluid 2 in patients, but with a significantly reduced incidence of days after treatment (FIG.3B), compared to a similar sample adverse events. taken before drug administration (FIG. 3A). The decrease in topoisomerase I levels directly seen by IHC in these cells at 48 Example 2 hours explains why much of the supercoiled DNA remains present in lanes of FIG. 3C with reactions from Day 2 lysates: Synthesis of 6,6'-Bis-(2-amino-2-carboxyleth i.e., there is much less enzyme available to act on these ylthio)-6,6'-dideoxy-3-cyclodextrin, 4 (CD-Bis Substrate molecules. Cys) 0729 Summary. In the phase I trial reported above, two dosing schedules, weekly x3 and every other week (bi 0733 weekly), were investigated. In the weekly x3 schedule the maximum tolerated dose was approximately 9 mg/m. Hema tologic toxicity and cystitis were the DLTs in this schedule. Scheme II Non-hematologic grade 3/4 adverse events included fatigue in 3 patients (25%), delayed onset hematuria/dysuria in 2 patients (17%), elevated CPK (creatine phosphokinase) in 1 HS OH patient (8%), and dehydration in 1 patient (8%), all of which were reversible. Cumulative bladder toxicity in some patients on this schedule was primarily observed post cycle one and O may have been related to the long terminal half-life of uncon jugated CPT, which is primarily cleared through the kidneys, I I leading to cumulative bladder irritation. Based on this obser Vation, it was decided to investigate a biweekly schedule as a O Y ( O strategy to reduce cumulative toxicity while maintaining dose delivery. --> r NH2 NH2 (0730. The biweekly schedule allowed for similar per cycle 4 plasma exposure to be achieved in patients but with a signifi cantly reduced incidence of adverse events. The only observed grade 3/4 adverse event was grade 3 neutropenia in 0734 167 mL of 0.1 M sodium carbonate buffer were one patient that was reversible. One dose reduction was degassed for 45 minutes in a 500 mL 2-neck round bottom required on this schedule. On this schedule, CRLX101 was flask equipped with a magnetic stir bar, a condenser and well tolerated without the toxicities normally associated with septum. To this solution were added 1.96 g (16.2 mmol) of camptothecin analogs, such as severe diarrhea and hemor L-cysteine and 10.0 g (73.8 mmol) of diiodo, deoxy-3-cyclo rhagic cystitis. The MTD on this schedule was determined to dextrin 2. The resulting Suspension was heated at a reflux be 15 mg/m. temperature for 4.5 h until the solution turned clear (color 0731 Pharmacokinetic analysis of CRLX101 was per less). The solution was then cooled to room temperature and formed after the first dose for all patients. Consistent with acidified to pH3 using 1 NHC1. The product was precipitated preclinical data, the pharmacokinetics of CRLX101 were by slow addition of acetone (3 times weight ratio of the characterized by a low volume of distribution and limited systemic clearance. Preclinical studies showed accumulation solution). This afforded 9.0 g crude material containing CD of CRLX101 in tumors and tissues of the reticuloendothelial biscysteine (90.0%), unreacted cyclodextrin, CD-mono-cys system such as liver and spleen. Increased release of active teine and cystine. The resulting Solid was subjected to anionic CPT from the conjugate was also observed in these tissues. exchange column chromatography (SuperQ65OM, Tosoh This study also confirmed that release kinetics of CPT were Bioscience) using a gradient elution of 0-0.4M ammonium Such that plasma levels of unconjugated CPT remained sig bicarbonate. All fractions were analyzed by HPLC. The nificantly below levels of conjugate at all times. After one desired fractions were combined and the solvent was reduced week, approximately 10% of the maximum recorded concen to 100 mL under vacuum. The final product was either pre tration of unconjugated CPT was still detected in plasma, cipitated by adding acetone or by adding methanol (3 times possibly leading to the cumulative toxicity observed on the weight ratio of the solution). 4 was obtained in 60-90% yield. weekly x3 schedule. On the biweekly schedule however, "H NMR (DO) & 5.08 (m, 7H, CD-2-CH), 3.79-3.94 (m, unconjugated CPT levels dropped below the limit of quanti 30H, CD-3,4-CH, CD-CH, Cys-CH), 3.49-3.62 (m. 14H, tation before the second dose, which may explain the lack of CD-5,6-CH), 2.92-3.30 (m, 4H, Cys-CH). 'C NMR (DO) urinary side effects on this schedule. & 172.3, 101.9, 83.9, 81.6, 81.5, 73.3, 72.2, 72.0, 60.7, 54.0, 0732. In general, CRLX101 was well tolerated and myelo 34.0, 30.6. ESI/MS (m/z): 1342 MI, 1364M+Na". Purity suppression was the DLT. Ten out of twenty-four patients of 4 was confirmed by HPLC US 2011/O160 159 A1 Jun. 30, 2011 62

Example 3 0736 t-Boc-glycine (0.9 g, 4.7 mmol) was dissolved in Synthesis of Gly-CPT (Structure 11) (Greenwald et 350 mL of anhydrous methylene chloride at room tempera al., Bioorg Med. Chem., 1998, 6, 551-562) ture, and to this solution were added DIPC (0.75 mL, 4.7 0735 mmol), DMAP (382 mg, 3.13 mmol) and camptothecin (0.55 g, 1.57 mmol) at 0°C. The reaction mixture was allowed to Scheme III warm to room temperature and left for 16 h. The solution was

washed with 0.1 NHCl, dried and evaporated under reduced pressure to yield a white solid, which was recrystallized from methanol to give camptothecin-20-ester of t-Boc-glycine: "H NMR (DMSO-d) 7.5-8.8 (m), 7.3 (s), 5.5 (s), 5.3 (s), 4 (m), 2.1 (m), 1.6 (s), 1.3 (d), 0.9 (t). Camptothecin-20-ester of t-Boc-glycine (0.595 g, 1.06 mmol) was dissolved in a mix ture of methylene chloride (7.5 mL) and TFA (7.5 mL) and stirred at room temperature for 1 h. Solvent was removed and the residue was recrystallized from methylene chloride and ether to give 0.45g of 11. HNMR (DMSO-d) 87.7-8.5 (m); 7.2 (s), 5.6 (s), 5.4 (s), 4.4 (m), 2.2 (m), 1.6 (d), 1.0 (t), "C NMR (DMSO-d) 868.6, 166.6, 156.5, 152.2, 147.9, 146.2, 144.3, 1319, 130.6, 129.7, 128.8, 128.6, 128.0, 127.8, 119.0, 95.0, 77.6, 66.6, 50.5, 47.9, 30.2, 15.9, 7.9. ESI/MS (m/z) expected 405; Found 406 (M+H).

Example 4

Synthesis and Characterization of CD-BisCys Peg3400 Copolymers 36 and their CPT Conjugates 37

0737 A. Synthesis and Characterization of CD-BisCys Peg3400 Copolymers 36

Scheme IWa

OH O

OH O O O O N O O o OH non-to- n-n n N OH O O O O OH HO O O HO O OH HO PEG-DiSPA (MW = 3400 kDa) DMSO/DIEA, TEA or DMAP

HO US 2011/O160 159 A1 Jun. 30, 2011

-continued

OH

OH O O O OH OH O HO OH O OH HO O HO O OH HO O O O H S OH HO N O NH OH S N-1- iii. pi O OH HO O O Ho O O OH O O O OH HO 36a-f Poly(CDDCys-PA-PEG) Abbr: PA = propanoicamide bond between PEG and CD

Scheme IVb OH

OH O O O OH OH HO O OH OH

O HO O HO O OH HO O O

HN S OH NH2 Comonomer BB S DMSO.DIEA HO O OH HO O O OH OH OH O HO O O O OH

HO US 2011/O160 159 A1 Jun. 30, 2011 64

-continued

O O O O N-- -N N1 ~~~);iii. O O O O PEG-DiSBA (MW = 3400 kDa) O N2N N1 O O N1(1-1 O N-1\o ls o1 N iii. 2N O PEG-DiBTC (MW =3400 kDa) or 'C' PEG-DiNPC (MW = 3400 kDa)

OH

OH O O O OH OH HO O HO OH

O HO O HO O OH HO O O S N OH H H N-R HO S pi O OH HO O O OH PH OH O HO O O O OH

HO 36g-i US 2011/O160 159 A1 Jun. 30, 2011 65

-continued ~~}- Poly(CDDCys-BA-PEG) O 1. N------36 R = O Poly(CDDCys-CB-PEG) O 1 N------36 R = O Poly(CDDCys-CB-PEG) Abbr: BA = butanoicamide bond; CB = carbamate bond

(0738 Synthesis of Poly(CDDCys-PA-PEG), 36a 4 (after no free cystine) gave higher MW polymer (36d-e) as com precipitation with acetone, 63 mg 0.047 mmol) and PEG pared to the less pure 4 that was obtained from the acetone DiSPA (MW 3400, 160 mg, 0.047 mmol) were dried under precipitation method (36a). Polymerization of 4 with PEG vacuum for 8 hours. Anhydrous DMSO (1.26 mL) was added DiSPA typically produced polymer yields greater than 90%. to the mixture under argon. After 10 minutes of stirring, 0741. 4 was polymerized with other activated monomers anhydrous diisopropylethylamine (DIEA, 19 uL., 2.3 eq.) was such as PEG-DiSBA, PEG-DiBTC, and PEG-DiNPC. Reac added under argon. The reaction mixture was stirred under tion of 4 with PEG-DiSBA gave polymer 36g with similar argon for 120 h. The polymer containing solution was dia linkages as 36a-f (amide bond, but one more —CH2 group lyzed using a 10,000 MWCO membrane (Spectra/Por 7) than 36a-fat the linker) with Mover 100 kDa, while reaction against water for 48 h and lyophilized to yield 196 mg 36a of 4 with PEG-DiBTC and PEG-DiNPC generated polymers (90%, Table 1). M=57.4 kDa, M-41.7 kDa, M/M-1.38. 36h and 36i, respectively, with connecting carbamate moiety H NMR (DO) & 5.08 (m, CD-2-H), 4.27 (m, Cys-CH), and M.'s over 50 kDa (Table 6). 2.72-3.76 (m, CD-3,4,5,6-CH, CD-CH, PEG-CH-), 2.44 (m, Cys-CH). TABLE 6 (0739 Synthesis of other poly(CDDCys-PA-PEG) (36b-f), Polymerization of 4 with difunctionalized PEG Poly(CDDCys-BA-PEG) (36g) Poly(CDDCys-CB-PEG) (36h-i) were achieved under polymerization condition simi Poly lar to that of 36a. Details for the polymerization conditions, PEG merization M. M. M. Yield monomer selection, polymer molecular weight, polydisper CDP Comonomer Base time (h) (kDa) (kDa) M, (%) 36a PEG-DSPA DIEA 120 57.4 41.7 1.38 90 sity and yields are listed in Table 6. 36g: "H NMR (DO) & 36b PEG-DSPA DMAP 120 54.2 38.1 1.42 91 5.10 (m, CD-2-H), 4.25-4.37 (m, Cys-CH), 2.72-3.86 (m, 36c PEG-DSPA TEA 120 57.4 42.6 1.35 91 CD-3,4,5,6-CH, CD-CH PEG-CH-), 2.21 (m, Cys-CH). 36d PEG-DSPA DIEA 120 93.6 58.O 148 96 36h-i: "H NMR (DO) & 5.05 (m, CD-2-H), 4.56 (m, Cys 36e PEG-DSPA DIEA 144 97.3 58.0 1.67 94 36f PEG-DSPA DIEA 2 35.3 25.6 1.38 95 CH), 2.70-3.93 (m, CD-3,4,5,6-CH, CD-CH, PEG-CH-), 36g PEG-DiSBA DIEA 120 114.7 77.9 1.47 96 2.38 (m, OCHCHCHC(O) NH ), 2.34 (m, Cys 36h PEG-DiBTC DIEA 120 67.6 39.4 1.47 95 CH), 1.90 (m, OCHCHCHC(O) NH-). 36i PEG-DNPC DIEA 120 86.5 57.2 1.51 96 0740. Addition of a non-nucleophilic organic base (such 4 was washed with acetone before polymerization. as DIEA) was essential for this polymerization as no viscosity 4 was washed with methanol before polymerization, changes of the polymerization solutions were observed after 48 hours if no base was added. When 2.3 eq. of DIEA were 0742 Polymers 36a-i are highly soluble in aqueous solu added, the Viscosity of the polymerization Solution increased tion. They can be easily dissolved in water or phosphate dramatically after 4-6 hours of reaction. DIEA deprotonates buffered saline (PBS) solution at concentrations of at least the amino groups of 4 to render them more nucleophilic for 200 mg/mL. Solubility of these polymers in aqueous solution coupling with PEG-DiSPA. There were essentially no differ at concentrations higher than 200 mg/mL was not attempted ences in the polymerizations if other bases, such as TEA or due to the high viscosity. These polymers were also soluble in DMAP, were used (36b-c, Table 6). Polymerization using 4 DMF, DMSO and methanol, slightly soluble in CHCN and recovered by the two different precipitation methods (acetone CHCl, but insoluble in THF and ethyl ether. and methanol) produced polymers with different MWs. 4 that 0743 Molecular Weight Control of CD Polymers 4 (after was purified by the methanol-precipitation method (contains precipitation with methanol) (56.2 mg, 0.0419 mmol) and US 2011/O160 159 A1 Jun. 30, 2011 66

PEG-DiSPA (147 mg, 0.0419 mmol) were dried under MWs of the precipitated polymers were determined as vacuum for 4-8 hours. To the mixture was added dry DMSO described above. (1.1L 2.2mL) eq) under was addedargon. under After argon. 10 minutes A portion stirring, of polymeriza- DIEA (16 B Synthesis of Poly(CDDYSPAPEG)-CPT Conjugates tion solution (150 uL) was removed and precipitated with (HGGG6, LGGG10, HG6, HGGG10). ether at selected times (2h, 18 h, 43 h, 70h, 168 hand 288 h). 0744)

Scheme W

He EDCNHS

IFA-NH O

36e or 36f Hs EDCNHS

TFA-NH2 O 12 US 2011/O160 159 A1 Jun. 30, 2011 67

-continued

OH HO

0745 Synthesis of Poly(CDDCys-PA-PEG)-GlyGlyGly 0747 Synthesis of Poly(CDDCys-PA-PEG)-Gly-CPT CPT (HGGG6) 36e (1.37 g., 0.30 mmol of repeat unit) was (HG6) Conjugation of 11 to 36e was performed in a manner dissolved in dry DMSO (136 mL). The mixture was stirred for similar to that used to produce HGGG6. The yield of HG6 10 minutes. 12 (419 mg 0.712 mmol, 2.36 eq), DIEA (0.092 was 83%. mL, 0.712 mmol. 2.36 eq), EDC (172 mg, 0.903 mmol. 3 eq), 0748 Synthesis of Poly(CDDCys-PA-PEG)-GlyGlyGly and NHS (76 mg, 0.662 mmol. 2.2 eq) were added to the CPT (HGGG10) 36e (1.5 g., 0.33 mmol of repeat unit) was polymer solution and stirred for ca. 15 hours. The polymer dissolved in dry DMSO (150 mL). The mixture was stirred for was precipitated with ethyl ether (1 L). The ether was poured 10 minutes. 12 (94.1 mg, 1.49 mmol. 4.5 eq), DIEA (0.258 out and the precipitate was washed with CHCN (3x100 mL). mL, 1.49 mmol. 4.5 eq), EDC (283 mg, 1.49 mmol. 4.5 eq), The precipitate was dissolved in water 600 mL. Some and NHS (113 mg 0.99 mmol. 3 eq) was added to the polymer solution and stirred for ca. 24 hours. Another portion of EDC insoluble solid was filtered through 0.2 Lum filters. The solu (142 mg, 0.75 mmol. 2.3 eq) and NHS (56 mg, 0.5 mmol. 1.5 tion was dialyzed using 25,000 MWCO membrane (Spectra/ eq) were added to the conjugation Solution. The polymer was Por 7) for 10 h at 10-15°C. in DI water. Dialysis water was stirred for an additional 22 hours. The workup procedure was changed every 60 minutes. The polymer-drug conjugate solu the same as that for the synthesis of HGGG6. The yield of tion was sterilized by passing it through 0.2 uM filters. The HGGG10 was 77%. solution was lyophilized to yield a yellow solid HGGG6 (1.42 0749. Determination of wt % CPT on the Conjugates g, 85% yield). Stock solutions of HGGG6, LGGG10, HG6 and HGGG10 0746 Synthesis of Poly(CDDCys-PA-PEG)-GlyGlyGly were prepared at a concentration of 10 mg/mL in DMSO. An CPT (LGGG10) Conjugation of 12 to 36f was performed in a aliquot of corresponding Stock solution was diluted to 100 manner similar to that used to produce HGGG6 except that ug/mL using 1 N NaOH. CPT was completely hydrolyzed in this conjugate was dialyzed with 10,000 MWCO membrane this basic solution and transformed to its carboxylate form (Spectra/Por 7) instead of with 25,000 MWCO membrane. within 2 hat room temperature. An aliquot of this solution The yield of LGGG10 was 83%. was diluted to 10 ug/mL using 8.5% HPO, and the CPT US 2011/O160 159 A1 Jun. 30, 2011

carboxylate form was transformed to its form. 30 uL 8.5% HPO prior to loading. After the acidified solution was of this solution was injected into the HPLC. The peak area loaded on the cartridge, the bed was washed with 3x1 mL of from the CPT lactone form was integrated and compared to a water. Released CPT and polymer conjugate were eluted with standard curve. 3x1 mL of a solution mixture of acetonitrile and potassium (0750 11 and 12 were conjugated to 36e or 36f(Table 2) phosphate buffer (pH 4.1) (60/40 V/v). The eluted solution using conventional coupling methods. Due to the instability was diluted to 5 mL total volume in a 5 mL volumetric flask. of the ester linker of 11 and 12 in aqueous solution, the 30 uL of such solution was injected into the HPLC. The peak conjugation was conducted in anhydrous DMSO under area from the CPT lactone form was integrated and compared argon. An organic base was required to deprotonate the TFA to a standard curve. salts of 11 and 12 to facilitate the coupling. For polymer conjugation with 12, the weight percent (wt %) drug loading 0754 Release of CPT from HGGG6 and HG6 in PBS was around 6-10%. The theoretical maximum drug loading is containing 4% human plasma (PBS/reconstituted human around 13% using PEG with MW of 3400 Da; maximum plasma Solution 96/4 (V/v)), in mouse plasma and in recon values can be increased by decreasing the MW of the PEG stituted human albumin (PBS solution) were performed in a segments. of all conjugates in water or PBS were manner similar to that described above for pure human more than 200 mg/mL (equivalent to a 12-20 mg. CPT/mL for plasma. 6-10 wt % drug loading, respectively). Details for the (0755. In PBS (1x, pH 7.4), the half-lives (t) for releasing HGGG6, LGGG10, HG6, and HGGG10 are summarized in CPT from HG6 and HGGG6 were 59 hand 32h, respectively. Table 7. The half-lives decreased to 25 hand 22 h, respectively, in the presence of 4% human plasma, and to 1.7 hand 1.6 h, respec TABLE 7 tively, in 100% human plasma (“HP) and 2.6 hand 2.2 h, respectively, in 100% mouse plasma (“MP). CPT release Properties of polymer-CPT conjugates. rates for both HG6 and HGGG6 in the presence of albumin M of parent CPT (“Alb’) or acetylcholinesterase (Ac Cho') were on the same Conjugate polymer (x10) M/M, Linker (wt %) order of magnitude as in PBS. In a buffer solution at a pH lower than PBS (pH 6.1) with or without the enzyme cathe HGGG6 97 1.7 triglycine 6.1 LGGG10 35 1.6 triglycine 10.2 psin B (active at pH 6.1), less than 50% of total conjugated HG6 97 1.7 glycine 6.8 CPT was released from both HG6 and HGGG6 for times up to HGGG10 97 1.7 triglycine 9.6 144 h (Table 8). Abbreviations; H = High M. polymer (97 kDa), L = Low M. polymer (35 kDa), GGG = triglycine linker, G = glycine linker, 6 = drug loading around 6 wt %, 10 = drug loading TABLE 8 around 10 wt.%. Polymer polydispersity as measured by light scattering techniques(26) Half-life (ta, in hour) of the release of CPT from HG6 and HGGG6 C. Release of CPT from HGGG6 and HG6 0751 Release of CPT in PBS HGGG6 and HG6 were Conju- 4% Ac pH 6.1 Cath B prepared at 1 mg/mL in PBS (1x, pH 7.4). A 100LL aliquot of gate PBS HP HP MP Alb, Chos buffer (pH 6.1) the solution was transferred to a 1.5 mL Eppendorf tube and HG6 S9 25 1.7 2.6 62 33 >144 >144 incubated at 37°C. The incubated samples were quenched at HGGG6 32 22 1.6 2.2 73 43 >144 >144 selected time intervals and storedat-80°C. until the analysis. to is defined as time (hours) for the release of half of the total conjugated CPT. Each solution was diluted with 8.5% HPO, to a 5 mL total Abbreviations: HP means human plasma, MP means mouse plasma, volume in a volumetric flask. 30 ul of such solution was pH 7.4 PBS 1x buffer. Reconstituted human plasma mixed with PBS (viv = 496). injected into the HPLC. The peak area from the CPT lactone *Reconstituted human plasma form was integrated and compared to a standard curve. Fresh mouse plasma (0752 Analysis for the release of CPT from HGGG6 and in reconstituted human albumin PBS buffer HG6 in PBS containing acetyl cholinesterase (an esterase, 3In the presence of acetylcholinesterase PBS solution (100 units/mL). 100 units/mL), in KHPO, buffer (pH 6.1, 0.1 M) and in the "pH 6.1 phosphate buffer (0.1M) KHPO buffer (pH 6.1, 0.1 M) containing cathepsin B (a pH 6.1 phosphate buffer in the presence of Cathepsin B cysteine proteinase, 200 uM, preactivated on ice for 30 min utes in this buffer containing 2 mM DTT and 1 mM EDTA) Release of CPT in Solution at Different pH. were performed in a manner similar to that described above for PBS alone. (0756 HGGG6 and HG6 were prepared at 1 mg/mL in (0753 Release of CPT in Human Plasma An aliquot of buffer solution with pHs ranging from acidic (pH=1.2) to HGGG6 and HG6 stock solution were diluted to give final basic (pH=13.1) and incubated at 37° C. for 24 h. An aliquot concentration of 0.5 mg/mL in PBS (1x, pH 7.4). This solu of each solution was diluted with 8.5% HPO to about 100 tion was added to a lyophilized powder of human plasma to ug/mL. 30 uL of such solution was injected into HPLC. The reconstitute 100% human plasma by the recommended peak area from the CPT lactone form was integrated and amount. The solution was divided into equal volume (250 uL) compared to a standard curve. to 1.5 mL Eppendorf tubes, incubated at 37°C., and stopped 0757. The pH of aqueous solution has a significant effect at selected time point. Samples were stored at -80° C. until on the CPT release rates from both HG6 and HGGG6. The the analysis. Samples were separated from plasma by Solid amounts of CPT released from HG6 and HGGG6 at 37° C. phase extraction columns The Solid phase extraction cartridge after 24 h in buffer solutions with pHs ranging from 1.1 to (Oasis HLB 1 cc cartridge from Waters) was pre-conditioned 13.1 are illustrated in FIG. 6. The glycinyl-CPT ester bonds of with 1 mL of acetonitrile and then with 1 mL of 8.5% HPO. both HG6 and HGGG6 were very stable in acidic pH (1.1 to before loading. Samples were acidified with equal volume of 6.4) as less than 7% of CPT were released in 24 h. US 2011/O160 159 A1 Jun. 30, 2011 69

0758 Methods for Increasing Drug Weight Percent Load 0761 Synthesis of polymer and drug conjugate 42 are ing same as 36, 37, and 38 (0759 Method I. Synthesis of CD-BisCys-Peg Copolymer 0762. While Scheme VI shows that the drug is attached at with a Short Peg Linkage and its GlyCPT Conjugate all available positions, not all positions may be reacted. Therefore, a particle comprising conjugates described above Example 5 may include a conjugate reacted at all positions available for Synthesis of CD-BisCys-Peg (Short PEG, e.g., attachment and particles that have less than all of the positions Peg200-Peg2000) and its CPT Conjugate 42 available for attachment containing the drug, e.g., the particle can include CPD reacted at one or none of the positions 0760 available for attachment. Thus, while Scheme VI depicts CPT at every point of attachment of each polymer Subunit, the CDP-CPT conjugate can have less than 2 CPT molecules Scheme VI attached to any given polymer subunit of the CDP. For O example, in one embodiment, the CDP-CPT conjugate includes several polymer Subunits and each of the polymer subunits can independently include two, one or no CPT O attached at each point of attachment of the polymer subunit. HN CriririO HN O In addition, the particles and compositions can include CDP CPT conjugates having two, one or no CPT attached to each Peg 200-2000 polymer subunit of the CDP-CPT conjugate and the conju gates can also include a mixture of CDP-CPT conjugates that can vary as to the number of CPTs attached at each point of attachment of the polymer Subunits of the conjugates in the particle or composition. 0763 Method II. Synthesis of CD-BisCys-Peg Copoly mer with multiple drug Molecules on each Loading Site. Example 6 Synthesis of CD-BisCys-Peg and its GluBis(Gly 42 CPT) Conjugate 43 0764

Scheme VII

score

PEG Mw = 3400 2 CPT per CD Maximum drug loading 13.4% US 2011/O160 159 A1 Jun. 30, 2011 70

-continued

l S

O

O

HN

O

43 PEG Mw = 3400 4. CPT per CD Maximum drug loading 25%

0765 36 and Glu-Bis(Gly-CPT) 17 are dissolved in Example 7 DMSO, EDC (3 eq), NHS (2.2 eq), and DIEA (2.2 eq) are added to the solution. CD-BisCys-Peg-GluBis(GlyCPT) 43 Synthesis and In vitro Analysis of CDP-Gly-SN-38 is precipitated with CHCN and washed with the same sol 0766 SN-38 was derivatized with the amino acid glycine vent until no free drug is detected using UV or TLC. 43 is at the 20-OH position as shown in Scheme VIII. Briefly, dried under high vacuum. While Scheme VII shows that the 20(S)-7-ethyl-10-hydroxycamptothecin (SN-38, 1.0 g, drug is attached at all available positions, not all positions mmol) was dissolved in a mixture of 70 mL dimethylforma may be reacted. Therefore, a particle comprising conjugates mide (DMF) and 30 mL pyridine. A solution of di-tert-butyl described above may include a conjugate reacted at all posi dicarbonate (0.83 g, 3.8 mmol) in 10 mL DMF was added and the mixture stirred at room temperature overnight (12 hours). tions available for attachment and particles that have less than The solvent was removed under vacuum to yield a yellow all of the positions available for attachment containing the solid and re-crystallized from boiling 2-propanol (75 mL) to drug, e.g., the particle can include CDP reacted at three, two, yield 200s)-10-tert-butoxycarbonyloxy-7-ethyl-camptoth one or none of the positions available for attachment. Thus, ecin (Boc-SN-38) as a yellow solid (0.6 g., 48% yield). while Scheme VII depicts CPT at every point of attachment of 0767 Boc-SN-38 (0.73 g, 1.5 mmol), N-(tertbutoxycar each polymer subunit, the CDP-CPT conjugate can have less bonyl)glycine (0.26g, 1.5 mmol) and 4-dimethylaminopyri than 4 CPT molecules attached to any given polymer subunit dine (DMAP, 0.18 g, 1.5 mmol) were dissolved in anhydrous of the CDP. For example, in one embodiment, the CDP-CPT methylene chloride (30 mL) and chilled to 0°C. 1,3-Diiso conjugate includes several polymer subunits and each of the propyl-carbodiimide (DIPC, 0.19 g, 1.5 mmol) was added, polymer Subunits can independently include four, three, two, the mixture stirred at 0°C. for 30 minutes followed by stirring one or no CPT attached at each point of attachment of the for 4 hours at room temperature. The mixture was diluted with polymer subunit. In addition, the particles and compositions methylene chloride to 100 mL, washed twice with an aqueous solution of 0.1N hydrochloric acid (25 mL), dried over mag can include CDP-CPT conjugates having four, three, two, one nesium sulfate and the solvent removed under vacuum. The or no CPT attached to each polymer subunit of the CDP-CPT resulting yellow solid was purified by flash chromatography conjugate and the conjugates can also include a mixture of in methylene chloride:acetone (9:1) followed by solvent CDP-CPT conjugates that can vary as to the number of CPTs removal under vacuum to yield 20-O-(N-(tert-butoxycarbo attached at each point of attachment of the polymer Subunits nyl) glycyl)-10-tert-butyoxycarbonyloxy-7-ethylcamptoth of the conjugates in the particle or composition. ecin (diBoc-Gly-SN-38, 640 mg, 67% yield). US 2011/O160 159 A1 Jun. 30, 2011 71

Scheme VIII: Derivatization of SN-38 to 20-O-(N-(tert butoxycarbonyl)glycyl)-10-tert-butyoxycarbonyloxy-7-eth ylcamptothecin (diBOC-Gly-SN-38)

> --k DMF:PYRIDINE (8:4) 2-Propanol Recrystallization 48% Yield

DMAP, DIPC/CH2Cl2 > ---, 1/2 h (a) 0° C. -->4 h (a) RT

CDP was synthesized as previously described (Cheng et al. Succinimide (14 mg., 0.12 mmol), and 1-ethyl-3-(3-dimethy (2003) Bioconjugate Chemistry 14(5):1007-1017). diBOC laminopropyl)carbodiimide (EDCI, 32 mg 0.17 mmol) were dissolved in dimethylformamide (10 mL) and stirred for 4 Gly-SN-38 (0.62g, 0.77 mmol) was deprotected in 15 mL of hours at room temperature. The polymer was precipitated by a 1:1 mixture of methylene chloride:trifluoroacetic acid addition of 50 mLacetone followed by 50 mL diethyl ether. (TFA) at room temperature for 1 hour. 20-O-trifluoroglycine Precipitate was centrifuged, washed twice with 20 mL 10-hydroxy-7-ethylcamptothecin (TFA-Gly-SN-38, 0.57 g. acetone each, and dissolved in water acidified to pH 3.0 with hydrochloric acid. Polymer solution was dialized for 24 hours 97% yield) was isolated as a yellow solid by precipitation against pH 3.0 water using a 25 kDa MWCO dialysis mem with ethanol (100 mL), followed by two washes with ethanol brane. The resulting solution was lyophilized to yield CDP (30 mL each), dissolution in methylene chloride and removal Gly-SN-38 (180 mg, 67% yield). The polymer was analyzed of solvent under vacuum. ESI/MS expected 449.4: Found for total and free SN-38 content by HPLC using SN-38 as a standard curve as previously described (Cheng et al. (2003) 471.66 (M+Na). Bioconjugate Chemistry 14(5):1007-1017). Total SN-38 0768 CDP-Gly-SN-38 (Poly-CD-PEG-Gly-SN-38, content was 7.66% w/w of which 97.4% was polymer bound. scheme IX) was synthesized as follows: CDP (270 mg, 0.056 Average particle size was determined by dynamic light scat mmol), TFA-Gly-SN-38 (70 mg 0.12 mmol), N-hydroxy tering to be 27.9 mm. US 2011/O160 159 A1 Jun. 30, 2011 72

Scheme IX: Structure of a subunit of CDP-Gly-SN-38

OH

0769 While Scheme IX shows that the drug is attached at at each point of attachment of the polymer subunits of the all available positions in the Subunit, not all positions may be conjugates in the particle or composition. reacted. Therefore, a particle comprising conjugates In vitro Evaluation of CDP-Gly-SN-38 described above may include a conjugate reacted at all posi (0770 CDP-Gly-SN-38 was evaluated in A2780 human tions available for attachment and particles that have less than ovarian cancer cell lines in vitro as follows: 0771. The human ovarian carcinoma A2780 cells were all of the positions available for attachment containing the obtained from the American Type Culture Collection. Cells drug, e.g., the particle can include CPD reacted at one or none were seeded in 96-well plates at a concentration of 5,000 cells of the positions available for attachment. Thus, while Scheme per well and grown in medium containing 10% fetal bovine IX depicts SN-38 at every point of attachment of each poly serum at 37°C. for 24h in a humidified 5% CO2 atmosphere. mer subunit, the CDP-SN-38 conjugate can have less than 2 The medium was replaced with fresh medium containing the SN-38 molecules attached to any given polymer subunit of test compound at concentrations ranging from 0.01 nmol/L to the CDP. For example, in one embodiment, the CDP-SN-38 1 umol/L. Triplicate wells per plate were treated at each conjugate includes several polymer subunits and each of the concentration. Controls were vehicle-treated cells and medium only blank. Plates were incubated at 37°C. for 72 h. polymer Subunits can independently include two, one or no MTS assay reagent was prepared by diluting CelTiter 96 Sn-38 attached at each point of attachment of the polymer AQueous One Solution (Promega) 5-fold into PBS/glucose Subunit. In addition, the particles and compositions can (4.5 g/L). Cell culture medium was aspirated and 100 uL of include CDP-SN-38 conjugates having two, one or no Sn-38 MTS reagent were added to each well. Plates were incubated attached to each polymer subunit of the CDP-Sn-38 conjugate at 37° C. for 1 h. The plates were shaken for 5 min and the and the conjugates can also include a mixture of CDP-Sn-38 absorbance was measured at 485 nm using a SPECTRAFluor conjugates that can vary as to the number of Sn-38s attached Plus plate reader (Tecan). The percentage of cell survival was US 2011/O160 159 A1 Jun. 30, 2011

calculated relative to untreated cells, and IC50s were esti 37. The method of claim 36, further comprising adminis mated from the graphs of log dose (nmol/L) versus '% cell tering one or more Subsequent administrations of the compo survival (GraphPad Prizm). sition. (0772. The results of this experiment are shown in Table 9 38. The method of claim 36, wherein the lung cancer is non below. Small cell lung cancer. 39. The method of claim 36, wherein lung cancer is squa TABLE 9 mous cell non Small cell lung cancer. 40. The method of claim 36, wherein the lung cancer is a IC50 of SN-38 and CDP-Gly-SN-38 on A2780 ovarian cells. Small cell lung cancer. 41. The method of claim 36, wherein the lung cancer is Compound IC50 (nM) Small cell lung cancer is a squamous Small cell lung cancer. SN-38 2.44 42. The method of claim 36, wherein the lung cancer is CDP-PEG-Gly-SN-38 7.22 locally advanced or metastatic lung cancer. 43. The method of claim 36, wherein the subject has a mutation in the EGFR gene. (0773) Other embodiments are in the claims. 44. The method of claim 36, wherein the lung cancer is sensitized to topoisomerase inhibitors. 1.-35. (canceled) 45. The method of claim 36, wherein the subject receives 36. A method of treating lung cancer in a subject, the radiation in combination with the administration of the com method comprising: administering a composition that com position. prises a CDP-camptothecin conjugate to a subject, wherein 46. The method of claim 36, wherein the lung cancer is the CDP-camptothecin conjugate has the following formula: refractory, relapsed or resistant to a platinum based agent or a taxane. 47. The method of claim 46, wherein the lung cancer is refractory, relapsed or resistant to carboplatin, cisplatin or oxaliplatin. 48. The method of claim 46, wherein the lung cancer is sc'.re-- refractory, relapsed or resistant to docetaxel, paclitaxel, laro taxel or cabazitaxel. 49. The method of claim 36, further comprising selecting wherein each D-L- is independently the subject for administration of the composition on the basis that the subject has increased KRAS expression levels as compared to the reference standard. 50. The method of claim 36, further comprising selecting the subject for administration of the composition on the basis that the subject has a mutation in the KRAS gene and/or the --> amino acid sequence of KRAS. 51. The method of claim 50, wherein the subject has a or absent, and each D is camptothecin, wherein at least one mutation at one or more of codon 12 of the KRAS gene, D-L- is codon 13 of the KRAS gene, and codon 61 of the KRAS gene. 52. The method of claim 36, wherein the subject has a mutation at one or more of codon 12 of the KRAS gene, codon 13 of the KRAS gene, and codon 61 of the KRAS gene. 53. The method of claim 43, wherein the subject has one or more of the following mutations: codon 719 of the EGFR --> -kn-r) gene, codon 746 of the EGFR gene, codon 747 of the EGFR gene, codon 748 of the EGFR gene, codon 749 of the EGFR gene, codon 750 of the EGFR gene, codon 858 of the EGFR has a Mw of 3.4 kDa or less, n is at least 4, and wherein the gene, a deletion in exon 19 of the EGFR gene, and an insert Subject has a mutation in the KRAS gene or has increased mutation at exon 20 of the EGFR gene. levels of KRAS expression as compared to a reference stan dard. c c c c c