throbber
UTILITY PATENT APPLICATION
`
`METHODS OF TREATING AND PREVENTING GRAFT VERSUS HOST DISEASE
`
`WSGR Docket No. 25922—885 .201
`
`Inventor(s):
`
`John C. BYRD
`
`Citizen of the United States of America, Residing at
`1950 Arlington Avenue
`Columbus, OH 43212
`
`Jason A. DUBOVSKY
`
`Citizen of the United States of America, Residing at
`2505 Avalon Place
`
`Columbus, OH 43219
`
`Nataraj an MUTHUSAMY
`Citizen of India, Residing at
`6129 Glenworth Court
`
`Galloway, OH 43119
`
`Amy J o JOHNSON
`Citizen of the United States of America, Residing at
`5396 Winters Run Road
`
`Dublin, OH 43016
`
`David MIKLOS
`
`Citizen of United States of America, Residing at
`875 Blake Wilbur Drive, MC 582
`
`Stanford, CA 94305
`
`Assignee:
`
`Pharmacyclics, Inc.
`
`995 East Arques Avenue
`
`Sunnyvale, CA 94085
`
`A Delaware corporation
`
`Entity:
`
`Large Business Concern
`
`\vfiR
`
`Wilson Sonsini Goodrich 5% Rosati
`PROHESSIONA i. fLflli‘P'JRA'I {ON
`
`650 Page Mill Road
`Palo Alto, CA 94304
`
`(650) 493—9300 (Main)
`
`(650) 493—6811 (Facsimile)
`
`Filed Electronically on: October 24, 2014
`
`SAN EX 1029, Page 01
`
`SAN EX 1029, Page 01
`
`

`

`WSGR Docket No. 25922—885201
`
`METHODS OF TREATING AND PREVENTING GRAFT VERSUS HOST DISEASE
`
`CROSS-REFERENCE
`
`[0001] This application claims the benefit of priority of US Provisional Application No.
`
`61/895,981, filed October 25, 2013; US Provisional Application No. 61/910,945, filed December 2,
`
`2013; US Provisional Application No. 61/973,173, filed March 31, 2014; and US Provisional
`
`Application No. 61/973,176 filed March 31, 2014, each of which is incorporated herein by reference.
`
`SEQUENCE LISTING
`
`[0001.1] The instant application contains a Sequence Listing which has been submitted in ASCII
`
`format via EFS —Web and is hereby incorporated by reference in its entirety. Said ASCII copy,
`
`created on October 20, 2014, is named 25922—885—20lSEQ.txt and is 633 bytes in size.
`
`BACKGROUND OF THE INVENTION
`
`[0002] Chronic graft versus host disease (cGVHD) is the most common long—term complication
`
`following allogeneic stem cell transplant (SCT), affecting 30—70% of patients who survive beyond
`
`the first 100 days. cGVHD and its associated immune deficiency have been identified as a leading
`
`cause of non—relapse mortality (NRM) in allogeneic SCT survivors. SCT survivors with cGVHD are
`
`4.7 times as likely to develop severe or life—threatening health conditions compared with healthy
`
`siblings, and patients with active cGVHD are more likely to report adverse general health, mental
`
`health, functional impairments, activity limitation, and pain than allo—SCT survivors with no history
`
`of cGVHD. Any organ system can be affected, and further morbidity is frequently caused by long—
`
`term exposure to the corticosteroids and calcineurin inhibitors required to treat the condition.
`
`SUMMARY OF THE INVENTION
`
`[0003] Disclosed herein, in some embodiments, are methods of preventing the occurrence of graft
`
`versus host disease (GVHD) or reducing the severity of GVHD occurrence in a patient requiring cell
`
`transplantation comprising administration of a therapeutically effective amount of an ACK inhibitor
`
`(e. g., an ITK or BTK inhibitor). In some embodiments, disclosed herein are methods of reducing the
`
`severity of GVHD occurrence in a patient requiring cell transplantation comprising administration of
`
`a therapeutically effective amount of an ACK inhibitor (e. g., an ITK or BTK inhibitor). In some
`
`embodiments the ACK inhibitor is a compound of Formula (A). In some embodiments, disclosed
`
`herein are methods of preventing the occurrence of graft versus host disease (GVHD) or reducing
`
`SAN EX 1029, Page 02
`
`SAN EX 1029, Page 02
`
`

`

`WSGR Docket No. 25922—885201
`
`the severity of GVHD occurrence in a patient requiring cell transplantation, comprising
`
`administration of a therapeutically effective amount of a compound of Formula (A) having the
`
`structure:
`
`R3\ 32
`N
`
`R1
`
`N \m%A
`
`N
`
`N
`R4
`
`Formula (A);
`
`wherein:
`
`Ais N;
`
`R1 is phenyl—O—phenyl or phenyl—S—phenyl;
`
`R2 and R3 are independently H;
`
`R4 is L3-X-L4-G, wherein,
`
`L3 is optional, and when present is a bond, optionally substituted or unsubstituted alkyl,
`
`optionally substituted or unsubstituted cycloalkyl, optionally substituted or unsubstituted alkenyl,
`
`optionally substituted or unsubstituted alkynyl;
`
`X is optional, and when present is a bond, —O—, —C(=O)—, —S—, —S(=O)—, —S(=O)2—, —NH—, —NR9—,
`
`—NHC(O)—, —C(O)NH—, —NR9C(O)—, —C(O)NR9—, —S(=O)2NH—, —NHS(=O)2—, —S(=O)2NR9—, —
`
`NRgS(=O)2—, —OC(O)NH—, —NHC(O)O—, —OC(O)NR9—, —NR9C(O)O—, —CH=NO—, —ON=CH—, —
`
`NR10C(O)NR10—, heteroaryl—, aryl—, —NR10C(=NR11)NR10—, —NR10C(=NR11)—, —C(=NR11)NR10—, -
`
`OC(=NR11)—, or —C(=NR11)O-;
`
`L4 is optional, and when present is a bond, substituted or unsubstituted alkyl, substituted or
`
`unsubstituted cycloalkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl,
`
`substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted
`
`heterocycle;
`
`or L3, X and L4 taken together form a nitrogen containing heterocyclic ring;
`
`q, ,0 R6
`0
`R6
`0
`/S /
`/
`EMR7 EMR LLL‘
`R8
`R8
`
`6 ,
`
`,
`
`R6
`E?
`/S\%\
`51‘
`R8
`
`R7
`
`R7
`
`,
`
`, or
`
`R6
`i 9
`_P%
`R26
`R8
`
`R7
`
`, wherein,
`
`G is
`
`-2-
`
`SAN EX 1029, Page 03
`
`SAN EX 1029, Page 03
`
`

`

`WSGR Docket No. 25922—885201
`
`R6, R7 and R3 are independently selected from among H, halogen, CN, OH, substituted or
`
`unsubstituted alkyl or substituted or unsubstituted heteroalkyl or substituted or unsubstituted
`
`cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl,
`
`substituted or unsubstituted heteroaryl;
`
`each R9 is independently selected from among H, substituted or unsubstituted lower alkyl,
`
`and substituted or unsubstituted lower cycloalkyl;
`
`each R10 is independently H, substituted or unsubstituted lower alkyl, or substituted or
`
`unsubstituted lower cycloalkyl; or
`
`two R10 groups can together form a 5—, 6—, 7—, or 8—membered heterocyclic ring; or
`
`R10 and R11 can together form a 5—, 6—, 7—, or 8—membered heterocyclic ring; or each R11 is
`
`independently selected from H or substituted or unsubstituted alkyl; or a pharmaceutically
`
`acceptable salt thereof. In some embodiments, L3, X and L4 taken together form a nitrogen
`
`containing heterocyclic ring. In some embodiments, the nitrogen containing heterocyclic ring is a
`
`piperidine group. In some embodiments, G is
`
`EMR7 911%
`R8
`or
`
`R6. In some embodiments,
`
`the compound of Formula (A) is l—[(3R)—3—[4—amino—3—(4—phenoxyphenyl)pyrazolo[3,4—d]pyrimidin—
`
`l—yl]piperidin—l—yl]prop—2—en—l—one. In some embodiments, the patient has cancer. In some
`
`embodiments, the patient has a hematological malignancy. In some embodiments, the patient has a
`
`relapsed or refractory hematological malignancy. In some embodiments, the patient has a B—cell
`
`malignancy. In some embodiments, the patient has a T—cell malignancy. In some embodiments, the
`
`patient has a leukemia, a lymphoma, or a myeloma. In some embodiments, the B—cell malignancy is
`
`a non—Hodgkin’s lymphoma. In some embodiments, the B—cell malignancy is chronic lymphocytic
`
`leukemia (CLL). In some embodiments, the B—cell malignancy is a relapsed or refractory B—cell
`
`malignancy. In some embodiments, the B—cell malignancy is a relapsed or refractory non—Hodgkin’s
`
`lymphoma. In some embodiments, the B—cell malignancy is a relapsed or refractory CLL. In some
`
`embodiments, the patient has high risk CLL. In some embodiments, the patient has a l7p
`
`chromosomal deletion. In some embodiments, the patient has 10%, 20%, 30%, 40%, 50%, 60%,
`
`70%, 80%, 90%, or greater CLL as determined by bone marrow biopsy. In some embodiments, the
`
`patient has received one or more prior anticancer agents. In some embodiments, the anticancer agent
`
`is selected from among alemtuzumab, bendamustine, bortezomib, CAL—101, chlorambucil,
`
`-3-
`
`SAN EX 1029, Page 04
`
`SAN EX 1029, Page 04
`
`

`

`WSGR Docket No. 25922—885201
`
`cyclophosphamide, dexamethasone, docetaxel, doxorubicin, endostatineverolimus, etoposide,
`
`fludarabine, fostamatinib, hydroxydaunorubicin, ibritumomab, ifosphamide, lenalidomide,
`
`mesalazine, ofatumumab, paclitaxel, pentostatin, prednisone, rituXimab, temsirolimus, thalidomide,
`
`tositumomab, vincristine, or a combination thereof. In some embodiments, the anticancer agent is
`
`rituXimab. In some embodiments, the anticancer agent is alemtuzumab. In some embodiments, the
`
`anticancer agent is fludarabine, cyclophosphamide, and rituXimab (FCR). In some embodiments, the
`
`anticancer agent is oxaliplatin, fludarabine, cytarabine, rituXimab (OFAR). In some embodiments,
`
`the amount of the ACK inhibitor compound (e. g., a compound of Formula (A)) prevents or reduces
`
`GVHD while maintaining a graft—versus—leukemia (GVL) reaction effective to reduce or eliminate
`
`the number of cancerous cells in the blood of the patient. In some embodiments, the cell
`
`transplantation is a hematopoietic cell transplantation. In some embodiments, the GVHD is acute
`
`GVHD. In some embodiments, the GVHD is chronic GVHD. In some embodiments, the GVHD is
`
`sclerodermatous GVHD. In some embodiments, the GVHD is steroid resistant GVHD. In some
`
`embodiments, the GVHD is cyclosporin—resistant GVHD. In some embodiments, the GVHD is
`
`refractory GVHD. In some embodiments, the GHVD is oral GVHD. In some embodiments, the oral
`
`GVHD is reticular oral GVHD. In some embodiments, the oral GVHD is erosive oral GVHD. In
`
`some embodiments, the oral GVHD is ulcerative oral GVHD. In some embodiments, the oral
`
`GVHD is GVHD of the oral cavity. In some embodiments, the oral GVHD is GVHD of the
`
`oropharyngeal region. In some embodiments, the oral GVHD is GVHD of the pharyngeal region. In
`
`some embodiments, the oral GVHD is GVHD of the esophageal region. In some embodiments, the
`
`oral GVHD is acute oral GVHD. In some embodiments, the oral GVHD is chronic oral GVHD. In
`
`some embodiments, the patient exhibits one or more symptoms of GVHD. In some embodiments,
`
`the patient has or will receive an allogeneic bone marrow or hematopoietic stem cell transplant. In
`
`some embodiments, the ACK inhibitor compound (e. g., a compound of Formula (A)) is administered
`
`concurrently with an allogeneic bone marrow or hematopoietic stem cell transplant. In some
`
`embodiments, the ACK inhibitor compound (e. g., a compound of Formula (A)) is administered prior
`
`to an allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments, the ACK
`
`inhibitor compound (e. g., a compound of Formula (A)) is administered subsequent to an allogeneic
`
`bone marrow or hematopoietic stem cell transplant. In some embodiments, the patient is a candidate
`
`for receiving HLA—mismatched hematopoietic stem cells. In some embodiments, the patient is a
`
`SAN EX 1029, Page 05
`
`SAN EX 1029, Page 05
`
`

`

`WSGR Docket No. 25922—885201
`
`candidate for receiving unrelated donor hematopoietic stem cells, umbilical vein hematopoietic stem
`
`cells, or peripheral blood stem cells. In some embodiments, the ACK inhibitor compound (e. g., a
`
`compound of Formula (A)) is administered orally. In some embodiments, the ACK inhibitor
`
`compound (e. g., a compound of Formula (A)) is administered at a dosage of between about 0.1
`
`mg/kg per day to about 100 mg/kg per day. In some embodiments, the ACK inhibitor compound
`
`(e. g., a compound of Formula (A)) is administered at a dosage of about 40 mg/day, about 140
`
`mg/day, about 280 mg/day, about 420 mg/day, about 560 mg/day, or about 840 mg/day. In some
`
`embodiments, the ACK inhibitor compound (e. g., a compound of Formula (A)) is administered in
`
`combination with other prophylactic agents. In some embodiments, the ACK inhibitor compound
`
`(e. g., a compound of Formula (A)) is administered from day l to about day 120 following allogeneic
`
`bone marrow or hematopoietic stem cell transplant. In some embodiments, the ACK inhibitor
`
`compound (e. g., a compound of Formula (A)) is administered from day l to about day 1000
`
`following allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments, the
`
`ACK inhibitor compound (e. g., a compound of Formula (A)) is administered in combination with
`
`one or more additional therapeutic agents. In some embodiments, the additional therapeutic agent is
`
`a corticosteroid. In some embodiments, the therapeutic agent is cyclosporine (CSA), mycophenolate
`
`mofetil (MMF) or a combination thereof. In some embodiments, the patient has or will receive a
`
`donor lymphocyte infusions (DLI).In some embodiments, the patient is administered one or more
`
`DLIs. In some embodiments, the patient is administered two or more DLIs. In some embodiments,
`
`the DLI comprises CD3+ lymphocytes. In some embodiments, the patient is administered one or
`
`more donor lymphocyte infusions (DLI) following an allogeneic bone marrow or hematopoietic stem
`
`cell transplant. In some embodiments, the ACK inhibitor compound (e. g., a compound of Formula
`
`(A)) is administered concurrently with a DLI following allogeneic bone marrow or hematopoietic
`
`stem cell transplant. In some embodiments, the ACK inhibitor compound (e. g., a compound of
`
`Formula (A)) is administered prior to a DLI following an allogeneic bone marrow or hematopoietic
`
`stem cell transplant. In some embodiments, the ACK inhibitor compound (e. g., a compound of
`
`Formula (A)) is administered following a DLI following an allogeneic bone marrow or
`
`hematopoietic stem cell transplant. In some embodiments, the ACK inhibitor compound (e. g., a
`
`compound of Formula (A)) is ibrutinib.
`
`SAN EX 1029, Page 06
`
`SAN EX 1029, Page 06
`
`

`

`WSGR Docket No. 25922—885201
`
`[0004] Disclosed herein, in some embodiments, are methods of treating a patient for alleviation of a
`
`bone marrow mediated disease, comprising administering to the patient allogeneic hematopoietic
`
`stem cells and/or allogeneic T—cells, and a therapeutically effective amount of an ACK inhibitor
`
`(e. g., an ITK or BTK inhibitor). In some embodiments, disclosed herein are methods of treating a
`
`patient for alleviation of a bone marrow mediated disease, with alleviation of consequently
`
`developed graft versus host disease (GVHD), comprising administering to the patient allogeneic
`
`hematopoietic stem cells and/or allogeneic T—cells, and a therapeutically effective amount of a
`
`compound of Formula (A):
`
`R3\ 32
`N
`
`R1
`
`N \m%A
`
`N
`
`N
`R4
`
`Formula (A);
`
`wherein:
`
`Ais N;
`
`R1 is phenyl—O—phenyl or phenyl—S—phenyl;
`
`R2 and R3 are independently H;
`
`R4 is L3-X-L4-G, wherein,
`
`L3 is optional, and when present is a bond, optionally substituted or unsubstituted alkyl,
`
`optionally substituted or unsubstituted cycloalkyl, optionally substituted or unsubstituted alkenyl,
`
`optionally substituted or unsubstituted alkynyl;
`
`X is optional, and when present is a bond, —O—, —C(=O)—, —S—, —S(=O)—, —S(=O)2—, —NH—, —NR9—,
`
`—NHC(O)—, —C(O)NH—, —NR9C(O)—, —C(O)NR9—, —S(=O)2NH—, —NHS(=O)2—, —S(=O)2NR9—, —
`
`NRgS(=O)2—, —OC(O)NH—, —NHC(O)O—, —OC(O)NR9—, —NR9C(O)O—, —CH=NO—, —ON=CH—, —
`
`NR10C(O)NR10—, heteroaryl—, aryl—, —NR10C(=NR11)NR10—, —NR10C(=NR11)—, —C(=NR11)NR10—, -
`
`OC(=NR11)—, or —C(=NR11)O-;
`
`L4 is optional, and when present is a bond, substituted or unsubstituted alkyl, substituted or
`
`unsubstituted cycloalkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl,
`
`substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted
`
`heterocycle;
`
`SAN EX 1029, Page 07
`
`SAN EX 1029, Page 07
`
`

`

`WSGR Docket No. 25922—885201
`
`or L3, X and L4 taken together form a nitrogen containing heterocyclic ring;
`
`0
`R6
`0
`/
`. a)? 7 HEMR6
`
`G is
`
`8
`
`,
`
`R
`
`Cos/’0 R6
`/ /
`IILL
`R
`
`8
`
`R
`
`7
`
`,
`
`,
`
`R6
`(IF?
`g
`R6
`(IS?
`/ % _ %\
`R20 R
`LLLL
`R
`7
`
`R
`
`I
`
`8
`
`, or
`
`8
`
`R
`
`7
`
`.
`
`, wherein,
`
`R6, R7 and R3 are independently selected from among H, halogen, CN, OH, substituted or
`
`unsubstituted alkyl or substituted or unsubstituted heteroalkyl or substituted or unsubstituted
`
`cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl,
`
`substituted or unsubstituted heteroaryl;
`
`each R9 is independently selected from among H, substituted or unsubstituted lower alkyl,
`
`and substituted or unsubstituted lower cycloalkyl;
`
`each R10 is independently H, substituted or unsubstituted lower alkyl, or substituted or
`
`unsubstituted lower cycloalkyl; or
`
`two R10 groups can together form a 5—, 6—, 7—, or 8—membered heterocyclic ring; or
`
`R10 and R11 can together form a 5—, 6—, 7—, or 8—membered heterocyclic ring; or
`
`each R11 is independently selected from H or substituted or unsubstituted alkyl; or a
`
`pharmaceutically acceptable salt thereof, is administered prior to, concurrently with, or following the
`
`allogeneic hematopoietic stem cells and/or allogeneic T—cells. In some embodiments, L3, X and L4
`
`taken together form a nitrogen containing heterocyclic ring. In some embodiments, the nitrogen
`
`containing heterocyclic ring is a piperidine group. In some embodiments, G is
`
`R8
`
`or
`
`571M
`
`R6. In some embodiments, the compound of Formula (A) is l—[(3R)—3—[4—amino—3—(4—
`
`phenoxyphenyl)pyrazolo [3 ,4—d] pyrimidin— l—yl] piperidin— l —yl] prop—2—en— 1 —one. In some
`
`embodiments, the patient has cancer. In some embodiments, the patient has a hematological
`
`malignancy. In some embodiments, the patient has a relapsed or refractory hematological
`
`malignancy. In some embodiments, the patient has a leukemia, a lymphoma, or a myeloma. In some
`
`embodiments, the patient has a B—cell malignancy. In some embodiments, the B—cell malignancy is a
`
`non—Hodgkin’s lymphoma. In some embodiments, the B—cell malignancy is chronic lymphocytic
`
`leukemia (CLL). In some embodiments, the B—cell malignancy is a relapsed or refractory B—cell
`
`malignancy. In some embodiments, the B—cell malignancy is a relapsed or refractory non—Hodgkin’s
`
`SAN EX 1029, Page 08
`
`SAN EX 1029, Page 08
`
`

`

`WSGR Docket No. 25922—885201
`
`lymphoma. In some embodiments, the B—cell malignancy is a relapsed or refractory CLL. In some
`
`embodiments, the patient has high risk CLL. In some embodiments, the patient has a l7p
`
`chromosomal deletion. In some embodiments, the patient has 10%, 20%, 30%, 40%, 50%, 60%,
`
`70%, 80%, 90%, or greater CLL as determined by bone marrow biopsy. In some embodiments, the
`
`patient has received one or more prior anticancer agents. In some embodiments, the anticancer agent
`
`is selected from among alemtuzumab, bendamustine, bortezomib, CAL—101, chlorambucil,
`
`cyclophosphamide, dexamethasone, docetaxel, doxorubicin, endostatineverolimus, etoposide,
`
`fludarabine, fostamatinib, hydroxydaunorubicin, ibritumomab, ifosphamide, lenalidomide,
`
`mesalazine, ofatumumab, paclitaxel, pentostatin, prednisone, rituXimab, temsirolimus, thalidomide,
`
`tositumomab, vincristine, or a combination thereof. In some embodiments, the anticancer agent is
`
`rituXimab. In some embodiments, the anticancer agent is alemtuzumab. In some embodiments, the
`
`anticancer agent is fludarabine, cyclophosphamide, and rituXimab (FCR). In some embodiments, the
`
`anticancer agent is oxaliplatin, fludarabine, cytarabine, rituXimab (OFAR). In some embodiments,
`
`the amount of the ACK inhibitor compound (e. g., a compound of Formula (A)) prevents or reduces
`
`GVHD while maintaining a graft—versus—leukemia (GVL) reaction effective to reduce or eliminate
`
`the number of cancerous cells in the blood of the patient. In some embodiments, the cell
`
`transplantation is a hematopoietic cell transplantation. In some embodiments, the GVHD is acute
`
`GVHD. In some embodiments, the GVHD is chronic GVHD. In some embodiments, the GVHD is
`
`sclerodermatous GVHD. In some embodiments, the GVHD is steroid resistant GVHD. In some
`
`embodiments, the GVHD is cyclosporin—resistant GVHD. In some embodiments, the GVHD is
`
`refractory GVHD. In some embodiments, the GHVD is oral GVHD. In some embodiments, the oral
`
`GVHD is reticular oral GVHD. In some embodiments, the oral GVHD is erosive oral GVHD. In
`
`some embodiments, the oral GVHD is ulcerative oral GVHD. In some embodiments, the oral
`
`GVHD is GVHD of the oral cavity. In some embodiments, the oral GVHD is GVHD of the
`
`oropharyngeal region. In some embodiments, the oral GVHD is GVHD of the pharyngeal region. In
`
`some embodiments, the oral GVHD is GVHD of the esophageal region. In some embodiments, the
`
`oral GVHD is acute oral GVHD. In some embodiments, the oral GVHD is chronic oral GVHD. In
`
`some embodiments, the patient exhibits one or more symptoms of GVHD. In some embodiments,
`
`the patient has or will receive an allogeneic bone marrow or hematopoietic stem cell transplant. In
`
`some embodiments, the ACK inhibitor compound (e. g., a compound of Formula (A)) is administered
`
`SAN EX 1029, Page 09
`
`SAN EX 1029, Page 09
`
`

`

`WSGR Docket No. 25922—885201
`
`concurrently with an allogeneic bone marrow or hematopoietic stem cell transplant. In some
`
`embodiments, the ACK inhibitor compound (e. g., a compound of Formula (A)) is administered prior
`
`to an allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments, the ACK
`
`inhibitor compound (e. g., a compound of Formula (A)) is administered subsequent to an allogeneic
`
`bone marrow or hematopoietic stem cell transplant. In some embodiments, the patient is a candidate
`
`for receiving HLA—mismatched hematopoietic stem cells. In some embodiments, the patient is a
`
`candidate for receiving unrelated donor hematopoietic stem cells, umbilical vein hematopoietic stem
`
`cells, or peripheral blood stem cells. In some embodiments, the ACK inhibitor compound (e. g., a
`
`compound of Formula (A)) is administered at a dosage of between about 0.1 mg/kg per day to about
`
`100 mg/kg per day. In some embodiments, the ACK inhibitor compound (e. g., a compound of
`
`Formula (A)) is administered at a dosage of about 40 mg/day, about 140 mg/day, about 280 mg/day,
`
`about 420 mg/day, about 560 mg/day, or about 840 mg/day. In some embodiments, the ACK
`
`inhibitor compound (e. g., a compound of Formula (A)) is administered orally. In some
`
`embodiments, the ACK inhibitor compound (e. g., a compound of Formula (A)) is administered in
`
`combination with additional therapeutic agents. In some embodiments, the additional therapeutic
`
`agent is a corticosteroid. In some embodiments, the additional therapeutic agent is cyclosporine
`
`(CSA), mycophenolate mofetil (MMF) or a combination thereof. In some embodiments, the ACK
`
`inhibitor compound (e. g., a compound of Formula (A)) is administered from day l to about day 120
`
`following allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments, the
`
`ACK inhibitor compound (e. g., a compound of Formula (A)) is administered from day l to about
`
`day 1000 following allogeneic bone marrow or hematopoietic stem cell transplant. In some
`
`embodiments, the patient has or will receive a donor lymphocyte infusion (DLI). In some
`
`embodiments, the patient has or will receive two or more donor lymphocyte infusions (DLI). In
`
`some embodiments, the patient is administered one or more donor lymphocyte infusions (DLI). In
`
`some embodiments, the DLI comprises CD3+ lymphocytes. In some embodiments, the patient is
`
`administered one or more donor lymphocyte infusions (DLI) following an allogeneic bone marrow
`
`or hematopoietic stem cell transplant. In some embodiments, the ACK inhibitor compound (e. g., a
`
`compound of Formula (A)) is administered concurrently with a DLI following allogeneic bone
`
`marrow or hematopoietic stem cell transplant. In some embodiments, the ACK inhibitor compound
`
`(e. g., a compound of Formula (A)) is administered prior to a DLI following an allogeneic bone
`
`SAN EX 1029, Page 010
`
`SAN EX 1029, Page 010
`
`

`

`WSGR Docket No. 25922—885201
`
`marrow or hematopoietic stem cell transplant. In some embodiments, the ACK inhibitor compound
`
`(e. g., a compound of Formula (A)) is administered following a DLI following an allogeneic bone
`
`marrow or hematopoietic stem cell transplant. In some embodiments, the ACK inhibitor compound
`
`(e. g., a compound of Formula (A)) is ibrutinib.
`
`[0005] In some embodiments, disclosed herein are methods of reducing the severity of GVHD
`
`occurrence in a patient requiring cell transplantation comprising administration of a therapeutically
`
`effective amount of ibrutinib (l—[(3R)—3—[4—amino—3—(4—phenoxyphenyl)pyrazolo[3,4—d]pyrimidin—l—
`
`yl]piperidin—l—yl]prop—2—en—l—one). In some embodiments, the patient has cancer. In some
`
`embodiments, the patient has a hematological malignancy. In some embodiments, the patient has a
`
`relapsed or refractory hematological malignancy. In some embodiments, the patient has a B—cell
`
`malignancy. In some embodiments, the patient has a T—cell malignancy. In some embodiments, the
`
`patient has a leukemia, a lymphoma, or a myeloma. In some embodiments, the B—cell malignancy is
`
`a non—Hodgkin’s lymphoma. In some embodiments, the B—cell malignancy is chronic lymphocytic
`
`leukemia (CLL). In some embodiments, the B—cell malignancy is a relapsed or refractory B—cell
`
`malignancy. In some embodiments, the B—cell malignancy is a relapsed or refractory non—Hodgkin’s
`
`lymphoma. In some embodiments, the B—cell malignancy is a relapsed or refractory CLL. In some
`
`embodiments, the patient has high risk CLL. In some embodiments, the patient has a l7p
`
`chromosomal deletion. In some embodiments, the patient has 10%, 20%, 30%, 40%, 50%, 60%,
`
`70%, 80%, 90%, or greater CLL as determined by bone marrow biopsy. In some embodiments, the
`
`patient has received one or more prior anticancer agents. In some embodiments, the anticancer agent
`
`is selected from among alemtuzumab, bendamustine, bortezomib, CAL—101, chlorambucil,
`
`cyclophosphamide, dexamethasone, docetaxel, doxorubicin, endostatineverolimus, etoposide,
`
`fludarabine, fostamatinib, hydroxydaunorubicin, ibritumomab, ifosphamide, lenalidomide,
`
`mesalazine, ofatumumab, paclitaxel, pentostatin, prednisone, rituXimab, temsirolimus, thalidomide,
`
`tositumomab, vincristine, or a combination thereof. In some embodiments, the anticancer agent is
`
`rituXimab. In some embodiments, the anticancer agent is alemtuzumab. In some embodiments, the
`
`anticancer agent is fludarabine, cyclophosphamide, and rituXimab (FCR). In some embodiments, the
`
`anticancer agent is oxaliplatin, fludarabine, cytarabine, rituXimab (OFAR). In some embodiments,
`
`the amount of ibrutinib prevents or reduces GVHD while maintaining a graft—versus—leukemia
`
`(GVL) reaction effective to reduce or eliminate the number of cancerous cells in the blood of the
`
`-10-
`
`SAN EX 1029, Page 011
`
`SAN EX 1029, Page 011
`
`

`

`WSGR Docket No. 25922—885201
`
`patient. In some embodiments, the cell transplantation is a hematopoietic cell transplantation. In
`
`some embodiments, the GVHD is acute GVHD. In some embodiments, the GVHD is chronic
`
`GVHD. In some embodiments, the GVHD is sclerodermatous GVHD. In some embodiments, the
`
`GVHD is steroid resistant GVHD. In some embodiments, the GVHD is cyclosporin—resistant
`
`GVHD. In some embodiments, the GVHD is refractory GVHD. In some embodiments, the GHVD
`
`is oral GVHD. In some embodiments, the oral GVHD is reticular oral GVHD. In some
`
`embodiments, the oral GVHD is erosive oral GVHD. In some embodiments, the oral GVHD is
`
`ulcerative oral GVHD. In some embodiments, the oral GVHD is GVHD of the oral cavity. In some
`
`embodiments, the oral GVHD is GVHD of the oropharyngeal region. In some embodiments, the oral
`
`GVHD is GVHD of the pharyngeal region. In some embodiments, the oral GVHD is GVHD of the
`
`esophageal region. In some embodiments, the oral GVHD is acute oral GVHD. In some
`
`embodiments, the oral GVHD is chronic oral GVHD. In some embodiments, the patient exhibits one
`
`or more symptoms of GVHD. In some embodiments, the patient has or will receive an allogeneic
`
`bone marrow or hematopoietic stem cell transplant. In some embodiments, the ibrutinib is
`
`administered concurrently with an allogeneic bone marrow or hematopoietic stem cell transplant. In
`
`some embodiments, the ibrutinib is administered prior to an allogeneic bone marrow or
`
`hematopoietic stem cell transplant. In some embodiments, the ibrutinib is administered subsequent to
`
`an allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments, the patient
`
`is a candidate for receiving HLA—mismatched hematopoietic stem cells. In some embodiments, the
`
`patient is a candidate for receiving unrelated donor hematopoietic stem cells, umbilical vein
`
`hematopoietic stem cells, or peripheral blood stem cells. In some embodiments, the ibrutinib is
`
`administered orally. In some embodiments, the ibrutinib is administered at a dosage of between
`
`about 0.1 mg/kg per day to about 100 mg/kg per day. In some embodiments, the ibrutinib is
`
`administered at a dosage of about 40 mg/day, about 140 mg/day, about 280 mg/day, about 420
`
`mg/day, about 560 mg/day, or about 840 mg/day. In some embodiments, the ibrutinib is
`
`administered in combination with other prophylactic agents. In some embodiments, the ibrutinib is
`
`administered from day l to about day 120 following allogeneic bone marrow or hematopoietic stem
`
`cell transplant. In some embodiments, the ibrutinib is administered from day l to about day 1000
`
`following allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments, the
`
`ibrutinib is administered in combination with one or more additional therapeutic agents. In some
`
`-11-
`
`SAN EX 1029, Page 012
`
`SAN EX 1029, Page 012
`
`

`

`WSGR Docket No. 25922—885201
`
`embodiments, the additional therapeutic agent is a corticosteroid. In some embodiments, the
`
`therapeutic agent is cyclosporine (CSA), mycophenolate mofetil (MMF) or a combination thereof. In
`
`some embodiments, the patient has or will receive a donor lymphocyte infusions (DLI).In some
`
`embodiments, the patient is administered one or more DLIs. In some embodiments, the patient is
`
`administered two or more DLIs. In some embodiments, the DLI comprises CD3+ lymphocytes. In
`
`some embodiments, the patient is administered one or more donor lymphocyte infusions (DLI)
`
`following an allogeneic bone marrow or hematopoietic stem cell transplant. In some embodiments,
`
`the ibrutinib is administered concurrently with a DLI following allogeneic bone marrow or
`
`hematopoietic stem cell transplant. In some embodiments, the ibrutinib is administered prior to a
`
`DLI following an allogeneic bone marrow or hematopoietic stem cell transplant. In some
`
`embodiments, the ibrutinib is administered following a DLI following an allogeneic bone marrow or
`
`hematopoietic stem cell transplant.
`
`INCORPORATION BY REFERENCE
`
`[0006] All publications, patents, and patent applications mentioned in this specification are herein
`
`incorporated by reference to the same extent as if each individual publication, patent, or patent
`
`application was specifically and individually indicated to be incorporated by reference.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`[0007] The novel features of the invention are set forth with particularity in the appended claims. A
`
`better understanding of the features and advantages of the present invention will be obtained by
`
`reference to the following detailed description that sets forth illustrative embodiments, in which the
`
`principles of the invention are utilized, and the accompanying drawings of which:
`
`[0008] FIG. 1 exemplifies that ibrutinib ameliorates cGVHD symptomatology after allotransplant.
`
`C57BL/6 mice were engrafted with LP/J bone marrow after 850 cGy lethal irradiation. 25 days post—
`
`transplant mice were randomly assigned to ibrutinib, vehicle, or cyclosporine groups. Panel A shows
`
`images showing external signs of cGVHD including alopecia, scleroderma, and fibrotic lesions at
`
`day 36 post—transplant. Ibrutinib treatment group displayed few external signs of cGVHD
`
`progression as compared to vehicle or cyclosporine groups. Panel B shows an analysis of cGVHD
`
`mouse groups using a physical scoring system adapted from Cooke et al., which incorporates weight,
`
`posture, coat condition, skin condition, and mobility. Scoring was conducted on day 36 post—
`
`transplantation. Panel C shows the LP/J—>C57BL/6 cGVHD scoring. Each category: coat condition,
`
`-12-
`
`SAN EX 1029, Page 013
`
`SAN EX 1029, Page 013
`
`

`

`WSGR Docket No. 25922—885201
`
`skin condition, weight, posture, mobility, and vitality are individually scored and summed to achieve
`
`an overall cGVHD condition score. Scores are taken by a consistent unbiased observer with no
`
`knowledge of treatment cohorts. Panel D provides images of cGVHD mouse groups at day 39 post—
`
`HSCT. Panel E provides images of H&E stained skin preparations of sclerodermatous skin lesions
`
`showing levels of dermal fibrosis, epidermal hyperplasia, serocellular crusting, erosion, and
`
`lymphohistiocytic infiltration, consistent with cGVHD.
`
`[0009] FIG. 2 exemplifies that Tregs are not inhibited by ibrutinib. Panel A provides a p

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket