COMBINATION THERAPY FOR CO-ADMINISTRATION OF MONOCLONAL ANTIBODIES

Information

  • Patent Application
  • 20170267753
  • Publication Number
    20170267753
  • Date Filed
    May 25, 2017
    7 years ago
  • Date Published
    September 21, 2017
    7 years ago
Abstract
Disclosed are methods for enhancing the efficacy of monoclonal antibody therapy, which entails co-administering a therapeutic monoclonal antibody, or a functional fragment thereof, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof. Also disclosed are methods of prolonging or increasing the time a monoclonal antibody remains in the circulation of a patient, which entails co-administering a therapeutic monoclonal antibody, or a functional fragment of the monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof, wherein the time the monoclonal antibody remains in the circulation (e.g., blood serum) of the patient is increased relative to the same regimen of administration of the monoclonal antibody but without the co-administration of the effective amount of colchicine and/or hydroxychloroquine. Further disclosed are therapeutic combinations, and kits containing the monoclonal antibodies and hydroxychloroquine and/or colchicine.
Description
BACKGROUND

Monoclonal antibodies have been developed for the treatment of a variety of conditions including autoimmune disorders, cancer, asthma, hypercholesterolemia and sepsis. The development of therapeutic monoclonal antibodies in these areas of medicine has progressed at a rapid pace. The number of new biologic agents that have been approved by the Food and Drug Administration (FDA) each year has quadrupled between 2004 and 2008. This relatively new class of medications has resulted in marked improvement in a number of patients with complex and potentially life-threatening conditions, and in some cases, they have replaced traditional small molecule pharmaceuticals as treatments of choice. These medications also accounted for approximately 17% of total global spending on medicines in 2016 with an overall market value of $200 billion.


An important influence on the utility of biologic therapy in an individual patient is the development of anti-drug antibodies (ADA). ADAs have been documented in patients receiving multiple doses of a variety monoclonal antibodies, including infliximab (IFX), a treatment for inflammatory bowel disease, rheumatoid arthritis, psoriatic arthritis and other autoimmune diseases. The development of ADAs to IFX, as well as other monoclonal antibodies, is associated with systemic reactions that can occur during or within a few days of drug infusion. When severe, they can require discontinuation of biologic therapies. In addition, a number of studies have shown that ADAs reduce the efficacy of biologic therapy. From a pharmacokinetic standpoint, ADAs have been shown to enhance the clearance of biologic medications. Strategies that have been developed to prevent ADA formation and their negative effect on the efficacy of biologic therapies include adherence to consistent timing of drug infusions or subcutaneous injection regimens, and the co-administration of oral immunomodulating medications such as thiopurines (azathioprine or its precursor agent 6-mercaptopurine), and methotrexate. Although studies have shown that this strategy of co-administration of the aforementioned immunomodulating agents reduces ADA production, rapid clearance of biologic agents and infusion reactions, patients on both classes of drugs may become further immunosuppressed, placing them at increased risk for opportunistic infections, tuberculosis, overwhelming fungal infections and a variety of cancers. In fact, hepatosplenic T-cell lymphoma, a rare, deadly cancer seen primarily in young males with Crohn's disease, has only been described in patients receiving monoclonal antibodies (MAb) against tumor necrosis factor alpha (TNF-α) in combination with thiopurine drugs.


It is therefore of high importance to develop methods to prevent ADA development in patients receiving biologic therapy that are safer than those that are currently in practice.


SUMMARY

Broadly, the present invention is based on the unexpected discovery that colchicine and hydroxychloroquine increase the time that a monoclonal antibody remains in the circulation or circulatory system (e.g., blood serum) of a patient by reducing the clearance of the monoclonal antibody from the body. The co-administration of colchicine and hydroxychloroquine and a monoclonal antibody thus increase the clearance time of monoclonal antibody from the body. Unlike known and conventional attempts to increase the effectiveness of monoclonal antibody therapies by preventing the formation of HACAs and other anti-drug antibodies (such as immunosuppression), the present invention may mitigate or even eliminate one of more of these drawbacks.


Accordingly, a first aspect of the present invention is directed to a method for enhancing the efficacy of monoclonal antibody therapy, which entails co-administering a therapeutic monoclonal antibody, or a functional fragment thereof, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof.


A related aspect of the present invention is directed to a method of prolonging or increasing the time a therapeutic monoclonal antibody remains in the circulation or circulatory system of a patient, which entails co-administering an effective amount of a therapeutic monoclonal antibody, or a functional fragment thereof, and an effective amount of colchicine, hydroxychloroquine, or a combination thereof, to a patient in need thereof. Clearance time of the monoclonal antibody from the circulation (e.g., blood serum) of the patient is increased relative to the same regimen of administration of the monoclonal antibody but without the co-administration of the effective amount of colchicine and/or hydroxychloroquine.


Another aspect of the present invention is directed to a therapeutic combination, which includes a therapeutically effective amount of a monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof.


A further aspect of the present invention is directed to a kit, which includes a therapeutic combination, which includes a therapeutically effective amount of a monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof. The kit may include both agents in a single dosage form or in separate dosage forms, in which case the respective dosage forms may be disposed in separate containers in the kit. The kit may further include printed instructions for using the therapeutic combination to practice the methods described herein.


In some embodiments of these aspects of the present invention, the monoclonal antibody is chronically administered (over a prolonged period of time) such as in the case of treatment of auto-immune diseases, e.g., monoclonal antibodies that target (and thus inhibit) TNF-α, such as adalimumab, certolizumab pegol, golimumab, and infliximab.


Hydroxychloroquine has a long history of use as an anti-malarial drug. Clinical studies have shown that hydroxychloroquine is not effective as a treatment for IBD, cancer, Clostridium infection, sepsis (except due to malaria), asthma or hyperchloresterolemia. Hydroxychloroquine is used as a disease-modifying anti-rheumatic drug in the treatment of rheumatoid arthritis and is commonly employed as treatment for systemic lupus erythematosis. Thus, hydroxychloroquine is effective treatment for several disorders. However, its specific use as combination therapy to enhance the efficacy of monoclonal antibody therapy has not been investigated. Without wishing to be bound to any particular theory, it is believed that hydroxychloroquine unexpectedly increases the time that a monoclonal antibody remains in the blood serum of a patient in one or more ways. It may decrease the clearance of the monoclonal antibody from the patient's system, for example, by inhibiting or reducing formation of antibodies such as human anti-chimeric antibodies (HACAs) or other anti-drug antibodies and decreasing the removal of the monoclonal antibody from the circulation. It is also believed that hydroxychloroquine raises lysosomal pH, which causes disruption of lysososmal function such as processing of antigens (such as monoclonal proteins) and antigen presentation to mononuclear cells.







DETAILED DESCRIPTION

Monoclonal antibodies (MAbs) that may be suitable for use in the present invention include human, humanized, chimeric and murine antibodies alike, as well as functional fragments thereof that bind the intended target, e.g., Fab fragments and Scfv fragments, and conjugated (e.g., pegylated MAbs and antibody-drug conjugates) and non-conjugated forms thereof. Representative examples of monoclonal antibodies are set forth in the table below, which FDA-approved MAbs.









TABLE 1







FDA Approved Monoclonal Antibodies











Antibody
Route
Type
Target
Indication





abciximab
intravenous
chimeric Fab
GPIIb/IIIa
Percutaneous coronary






intervention


adalimumab
subcutaneous
fully human
TNF
Rheumatoid arthritis


adalimumab-
subcutaneous
fully
TNF
Rheumatoid arthritis


atto

human, biosimilar

Juvenile idiopathic arthritis






Psoriatic arthritis






Ankylosing spondylitis






Crohn's disease






Ulcerative colitis






Plaque psoriasis


ado-
intravenous
humanized, antibody-
HER2
Metastatic breast cancer


trastuzumab

drug conjugate


emtansine


alemtuzumab
intravenous
humanized
CD52
B-cell chronic lymphocytic






leukemia


alirocumab
subcutaneous
fully human
PCSK9
Heterozygous familial






hypercholesterolemia






Refractory hypercholesterolemia


atezolizumab
intravenous
humanized
PD-L1
Urothelial carcinoma


atezolizumab
intravenous
humanized
PD-L1
Urothelial carcinoma






Metastatic non-small cell lung






cancer


avelumab
intravenous
fully human
PD-L1
Metastatic Merkel cell






carcinoma


basiliximab
intravenous
chimeric
IL2RA
Prophylaxis of acute organ






rejection in renal transplant


belimumab
intravenous
fully human
BLyS
Systemic lupus erythematosus


bevacizumab
intravenous
humanized
VEGF
Metastatic colorectal cancer


bezlotoxumab
intravenous
fully human

Clostridium

Prevent recurrence






difficile toxin B

of Clostridium difficile infection


blinatumomab
intravenous
mouse, bispecific
CD19
Precursor B-cell acute






lymphoblastic leukemia


brentuximab
intravenous
chimeric, antibody-
CD30
Hodgkin lymphoma


vedotin

drug conjugate

Anaplastic large-cell lymphoma


brodalumab
subcutaneous
chimeric
IL17RA
Plaque psoriasis


canakinumab
subcutaneous
fully human
IL1B
Cryopyrin-associated periodic






syndrome


capromab
intravenous
murine, radiolabeled
PSMA
Diagnostic imaging agent in


pendetide



newly-diagnosed prostate






cancer or post-prostatectomy


certolizumab
subcutaneous
humanized
TNF
Crohn's disease


pegol


cetuximab
intravenous
chimeric
EGFR
Metastatic colorectal carcinoma


daclizumab
intravenous
humanized
IL2RA
Prophylaxis of acute organ






rejection in renal transplant


daclizumab
subcutaneous
humanized
IL2R
Multiple sclerosis


daratumumab
intravenous
fully human
CD38
Multiple myeloma


denosumab
subcutaneous
fully human
RANKL
Postmenopausal women






with osteoporosis


dinutuximab
intravenous
chimeric
GD2
Pediatric high-






risk neuroblastoma


dupilumab
subcutaneous
fully human
IL4RA
Atopic dermatitis


durvalumab
intravenous
fully human
PD-L1
Urothelial carcinoma


eculizumab
intravenous
humanized
Complement
Paroxysmal nocturnal





component 5
hemoglobinuria


elotuzumab
intravenous
humanized
SLAMF7
Multiple myeloma


evolocumab
subcutaneous
fully human
PCSK9
Heterozygous familial






hypercholesterolemia






Refractory hypercholesterolemia


golimumab
subcutaneous
fully human
TNF
Rheumatoid arthritis






Psoriatic arthritis






Ankylosing spondylitis


golimumab
intravenous
fully human
TNF
Rheumatoid arthritis


ibritumomab
intravenous
murine,
CD20
Relapsed or refractory low-


tiuxetan

radioimmunotherapy

grade, follicular, or transformed






B-cell non-Hodgkin's lymphoma


idarucizumab
intravenous
humanized Fab
dabigatran
Emergency reversal of






anticoagulant dabigatran


infliximab
intravenous
chimeric
TNF alpha
Crohn's disease


inflixmab-abda
intravenous
chimeric, biosimilar
TNF
Crohn's disease






Ulcerative colitis






Rheumatoid arthritis






Ankylosing spondylitis






Psoriatic arthritis






Plaque psoriasis


infliximab-
intravenous
chimeric, biosimilar
TNF
Crohn's disease


dyyb



Ulcerative colitis






Rheumatoid arthritis






Ankylosing spondylitis






Psoriatic arthritis






Plaque psoriasis


ipilimumab
intravenous
fully human
CTLA-4
Metastatic melanoma


ixekizumab
subcutaneous
humanized
IL17A
Plaque psoriasis


mepolizumab
subcutaneous
humanized
IL5
Severe asthma


natalizumab
intravenous
humanized
alpha-4 integrin
Multiple sclerosis


necitumumab
intravenous
fully human
EGFR
Metastatic squamous non-small






cell lung carcinoma


nivolumab
intravenous
fully human
PD-1
Metastatic melanoma


nivolumab
intravenous
fully human
PD-1
Metastatic squamous non-small






cell lung carcinoma


obiltoxaximab
intravenous
chimeric
Protective
Inhalational anthrax





antigen of





the Anthrax





toxin


obinutuzumab
intravenous
humanized
CD20
Chronic lymphocytic leukemia


ocrelizumab
intravenous
humanized
CD20
Multiple sclerosis


ofatumumab
intravenous
fully human
CD20
Chronic lymphocytic leukemia


olaratumab
intravenous
fully human
PDGFRA
Soft tissue sarcoma


omalizumab
intravenous
humanized
IgE
Moderate to severe






persistent asthma


palivizumab
intramuscular
humanized
F protein
Respiratory syncytial virus





of RSV


panitumumab
intravenous
fully human
EGFR
Metastatic colorectal cancer


pembrolizumab
intravenous
humanized
PD-1
Metastatic melanoma


pertuzumab
intravenous
humanized
HER2
Metastatic breast cancer


ramucirumab
intravenous
fully human
VEGFR2
Gastric cancer


ranibizumab
intravitreal
humanized
VEGFR1
Wet age-related macular



injection

VEGFR2
degeneration


raxibacumab
intravenous
fully human
Protective
Inhalational anthrax





antigen





of Bacillus






anthracis



reslizumab
intravenous
humanized
IL5
Severe asthma


rituximab
intravenous
chimeric
CD20
B-cell non-Hodgkin's lymphoma


secukinumab
subcutaneous
fully human
IL17A
Plaque psoriasis


siltuximab
intravenous
chimeric
IL6
Multicentric Castleman's disease


tocilizumab
intravenous
humanized
IL6R
Rheumatoid arthritis


tocilizumab
intravenous
humanized
IL6R
Rheumatoid arthritis



subcutaneous


Polyarticular juvenile idiopathic






arthritis






Systemic juvenile idiopathic






arthritis


trastuzumab
intravenous
humanized
HER2
Metastatic breast cancer


ustekinumab
subcutaneous
fully human
IL12
Plaque psoriasis





IL23


ustekinumab
subcutaneous
fully human
IL12
Plaque psoriasis



intravenous

IL23
Psoriatic arthritis






Crohn's disease


vedolizumab
intravenous
humanized
integrin receptor
Ulcerative colitis






Crohn's disease









Other representative examples of monoclonal antibodies that may be suitable for use in the present invention are listed in Table 2.









TABLE 2







Therapeutic Monoclonal Antibodies











Name
Type
Source
Target
Use





3F8
mab
mouse
GD2 ganglioside
neuroblastoma


8H9
mab
mouse
B7-H3
neuroblastoma, sarcoma,






metastatic brain cancers


Abagovomab
mab
mouse
CA-125 (imitation)
ovarian cancer


Abciximab
Fab
chimeric
CD41 (integrin alpha-IIb)
platelet aggregation inhibitor


Abituzumab
mab
humanized
CD51
cancer


Abrilumab
mab
human
integrin α4β7
inflammatory bowel






disease, ulcerative






colitis, Crohn's disease


Actoxumab
mab
human

Clostridium difficile


Clostridium difficile colitis



Adalimumab
mab
human
TNF-α
Rheumatoid arthritis, Crohn's






Disease,






Plaque Psoriasis, Psoriatic






Arthritis, Ankylosing






Spondylitis, Juvenile






Idiopathic






Arthritis, Hemolytic disease






of the newborn


Adecatumumab
mab
human
EpCAM
prostate and breast cancer


Aducanumab
mab
human
beta-amyloid
Alzheimer's disease


Afelimomab
F(ab′)2
mouse
TNF-α
sepsis


Afutuzumab
mab
humanized
CD20
lymphoma


Alacizumab pegol
F(ab′)2
humanized
VEGFR2
cancer


Alemtuzumab
mab
humanized
CD52
Multiple sclerosis


Alirocumab
mab
human
PCSK9
hypercholesterolemia


Altumomab
mab
mouse
CEA
colorectal cancer (diagnosis)


pentetate


Amatuximab
mab
chimeric
mesothelin
cancer


Anatumomab
Fab
mouse
TAG-72
non-small cell lung


mafenatox



carcinoma


Anetumab
mab
human
MSLN
cancer


ravtansine


Anifrolumab
mab
human
interferon α/β receptor
systemic lupus






erythematosus


Anrukinzumab
mab
humanized
IL-13
asthma


(=IMA-638)


Arcitumomab
Fab′
mouse
CEA
gastrointestinal cancers






(diagnosis)


Ascrinvacumab
mab
human
activin receptor-like kinase
cancer





1


Aselizumab
mab
humanized
L-selectin (CD62L)
severely injured patients


Atezolizumab
mab
humanized
CD274
cancer


Atlizumab
mab
humanized
IL-6 receptor
rheumatoid arthritis


(=tocilizumab)


Atorolimumab
mab
human
Rhesus factor
hemolytic disease of the






newborn


Bapineuzumab
mab
humanized
beta amyloid
Alzheimer's disease


Basiliximab
mab
chimeric
CD25 (α chain of IL-
prevention of





2 receptor)
organ transplant rejections


Bavituximab
mab
chimeric
phosphatidylserine
cancer, viral infections


Bectumomab
Fab′
mouse
CD22
non-Hodgkin's






lymphoma (detection)


Belimumab
mab
human
BAFF
non-Hodgkin lymphoma etc.


Benralizumab
mab
humanized
CD125
asthma


Bertilimumab
mab
human
CCL11 (eotaxin-1)
severe allergic disorders


Besilesomab
mab
mouse
CEA-related antigen
inflammatory lesions and






metastases (detection)


Bevacizumab
mab
humanized
VEGF-A
metastatic






cancer, retinopathy of






prematurity


Bezlotoxumab
mab
human

Clostridium difficile


Clostridium difficile colitis



Biciromab
Fab′
mouse
fibrin II, beta chain
thromboembolism






(diagnosis)


Bimagrumab
mab
human
ACVR2B
myostatin inhibitor


Bivatuzumab
mab
humanized
CD44 v6
squamous cell carcinoma


mertansine


Blinatumomab
BiTE
mouse
CD19
pre-B ALL (CD 19+)


Blosozumab
mab
humanized
SOST
osteoporosis


Bococizumab
mab
humanized
neural apoptosis-regulated
dyslipidemia





proteinase 1


Brazikumab
mab
human
IL23
Crohn's disease


Brentuximab
mab
chimeric
CD30 (TNFRSF8)
hematologic cancers


vedotin


Briakinumab
mab
human
IL-12, IL-23
psoriasis, rheumatoid






arthritis, inflammatory bowel






diseases, multiple sclerosis


Brodalumab
mab
human
IL-17
inflammatory diseases


Brolucizumab
mab
humanized
VEGFA
wet age-related macular






degeneration


Brontictuzumab
mab
humanized
Notch 1
cancer


Burosumab
mab
human
FGF23
X-linked hypophosphatemia


Cantuzumab
mab
humanized
mucin CanAg
colorectal cancer etc.


mertansine


Cantuzumab
mab
humanized
MUC1
cancers


ravtansine


Caplacizumab
mab
humanized
VWF
thrombotic






thrombocytopenic






purpura, thrombosis


Capromab
mab
mouse
prostatic carcinoma cells
prostate cancer (detection)


pendetide


Carlumab
mab
human
MCP-1
oncology/immune






indications


Catumaxomab
3funct
rat/mouse hybrid
EpCAM, CD3
ovarian cancer,






malignant ascites, gastric






cancer


cBR96-doxorubicin
mab
humanized
Lewis-Y antigen
cancer


immunoconjugate


Cedelizumab
mab
humanized
CD4
prevention of organ






transplant rejections,






treatment of autoimmune






diseases


Certolizumab pegol
Fab′
humanized
TNF-α
Crohn's disease Rheumatoid






arthritis axial






spondyloarthritis psoriasis






arthritis


Cetuximab
mab
chimeric
EGFR
metastatic colorectal






cancer and head and neck






cancer


Ch.14.18
mab
chimeric
GD2 ganglioside
neuroblastoma


Citatuzumab
Fab
humanized
EpCAM
ovarian cancer and other


bogatox



solid tumors


Cixutumumab
mab
human
IGF-1 receptor (CD221)
solid tumors


Clazakizumab
mab
humanized

Oryctolagus cuniculus

rheumatoid arthritis


Clenoliximab
mab
chimeric
CD4
rheumatoid arthritis


Clivatuzumab
mab
humanized
MUC1
pancreatic cancer


tetraxetan


Codrituzumab
mab
humanized
glypican 3
cancer


Coltuximab
mab
chimeric
CD19
cancer


ravtansine


Conatumumab
mab
human
TRAIL-R2
cancer


Concizumab
mab
humanized
TFPI
bleeding


CR6261
mab
human
Influenza A hemagglutinin
infectious disease/influenza A


Crenezumab
mab
humanized
1-40-β-amyloid
Alzheimer's disease


Crotedumab
mab
human
GCGR
diabetes


Dacetuzumab
mab
humanized
CD40
hematologic cancers


Daclizumab
mab
humanized
CD25 (α chain of IL-
prevention of organ





2 receptor)
transplant rejections


Dalotuzumab
mab
humanized
IGF-1 receptor (CD221)
cancer etc.


Daratumumab
mab
human
CD38 (cyclic ADP ribose
cancer





hydrolase)


Demcizumab
mab
humanized
DLL4
cancer


Denintuzumab
mab
humanized
CD19
cancer


mafodotin


Denosumab
mab
human
RANKL
osteoporosis, bone






metastases etc.


Depatuxizumab
mab
chimeric/humanized
EGFR
cancer


mafodotin


Derlotuximab biotin
mab
chimeric
histone complex
recurrent glioblastoma






multiform


Detumomab
mab
mouse
B-lymphoma cell
lymphoma


Dinutuximab
mab
chimeric
GD2 ganglioside
neuroblastoma


Diridavumab
mab
human
hemagglutinin
influenza A


Domagrozumab
mab
humanized
GDF-8
Duchenne muscular






dystrophy


Drozitumab
mab
human
DR5
cancer etc.


Duligotumab
mab
human
ERBB3 (HER3)
testicular cancer


Dupilumab
mab
human
IL4
atopic diseases


Durvalumab
mab
human
CD274
cancer


Dusigitumab
mab
human
ILGF2
cancer


Ecromeximab
mab
chimeric
GD3 ganglioside
malignant melanoma


Eculizumab
mab
humanized
C5
paroxysmal nocturnal






hemoglobinuria, atypical






HUS


Edobacomab
mab
mouse
endotoxin
sepsis caused by Gram-






negative bacteria


Edrecolomab
mab
mouse
EpCAM
colorectal carcinoma


Efalizumab
mab
humanized
LFA-1 (CD11a)
psoriasis (blocks T-






cell migration)


Efungumab
scFv
human
Hsp90
invasive Candida infection


Eldelumab
mab
human
interferon gamma-induced
Crohn's disease, ulcerative





protein
colitis


Elgemtumab
mab
human
ERBB3 (HER3)
cancer


Elotuzumab
mab
humanized
SLAMF7
multiple myeloma


Emactuzumab
mab
humanized
CSF1R
cancer


Emibetuzumab
mab
humanized
HHGFR
cancer


Emicizumab
mab
humanized
activated F9, F10
haemophilia A


Enavatuzumab
mab
humanized
TWEAK receptor
cancer etc.


Enfortumab vedotin
mab
human
AGS-22M6
cancer expressing Nectin-4


Enoblituzumab
mab
humanized
CD276
cancer


Enokizumab
mab
humanized
IL9
asthma


Ensituximab
mab
chimeric
5AC
cancer


Epratuzumab
mab
humanized
CD22
cancer, SLE


Erenumab
mab
human
CGRP
migraine


Erlizumab
F(ab′)2
humanized
ITGB2 (CD18)
heart






attack, stroke, traumatic






shock


Ertumaxomab
3funct
rat/mouse hybrid
HER2/neu, CD3
breast cancer etc.


Etaracizumab
mab
humanized
integrin αvβ3
melanoma, prostate






cancer, ovarian cancer etc.


Etrolizumab
mab
humanized
integrin α7 β7
inflammatory bowel disease


Evinacumab
mab
human
angiopoietin 3
dyslipidemia


Evolocumab
mab
human
PCSK9
hypercholesterolemia


Exbivirumab
mab
human
hepatitis B surface antigen
hepatitis B


Fanolesomab
mab
mouse
CD15
appendicitis (diagnosis)


Farletuzumab
mab
humanized
folate receptor 1
ovarian cancer


Fasinumab
mab
human
HNGF
acute sciatic pain


FBTA05
3funct
rat/mouse hybrid
CD20
chronic lymphocytic






leukaemia


Felvizumab
mab
humanized
respiratory syncytial virus
respiratory syncytial virus






infection


Fezakinumab
mab
human
IL-22
rheumatoid






arthritis, psoriasis


Ficlatuzumab
mab
humanized
HGF
cancer etc.


Figitumumab
mab
human
IGF-1 receptor (CD221)
adrenocortical






carcinoma, non-small cell






lung carcinoma etc.


Flanvotumab
mab
human
TYRP1(glycoprotein 75)
melanoma


Fletikumab
mab
human
IL 20
rheumatoid arthritis


Fontolizumab
mab
humanized
IFN-γ
Crohn's disease etc.


Foravirumab
mab
human
rabies virus glycoprotein
rabies (prophylaxis)


Fresolimumab
mab
human
TGF-β
idiopathic pulmonary






fibrosis, focal segmental






glomerulosclerosis, cancer


Fulranumab
mab
human
NGF
pain


Futuximab
mab
chimeric
EGFR
cancer


Galcanezumab
mab
humanized
calcitonin
migraine


Galiximab
mab
chimeric
CD80
B-cell lymphoma


Ganitumab
mab
human
IGF-1 receptor (CD221)
cancer


Gantenerumab
mab
human
beta amyloid
Alzheimer's disease


Gavilimomab
mab
mouse
CD147 (basigin)
graft versus host disease


Gemtuzumab
mab
humanized
CD33
acute myelogenous leukemia


ozogamicin


Gevokizumab
mab
humanized
IL-1β
diabetes etc.


Girentuximab
mab
chimeric
carbonic anhydrase 9 (CA-
clear cell renal cell





IX)
carcinoma


Glembatumumab
mab
human
GPNMB
melanoma, breast cancer


vedotin


Golimumab
mab
human
TNF-α
rheumatoid arthritis,






psoriatic arthritis, ankylosing






spondylitis


Gomiliximab
mab
chimeric
CD23 (IgE receptor)
allergic asthma


Guselkumab
mab
human
IL23
psoriasis


Ibalizumab
mab
humanized
CD4
HIV infection


Ibritumomab
mab
mouse
CD20
non-Hodgkin's lymphoma


tiuxetan


Icrucumab
mab
human
VEGFR-1
cancer etc.


Idarucizumab
mab
humanized
dabigatran
reversal of anticoagulant






effects of dabigatran


Igovomab
F(ab′)2
mouse
CA-125
ovarian cancer (diagnosis)


IMAB362
mab
human
CLDN18.2
gastrointestinal






adenocarcinomas and






pancreatic tumor


Imalumab
mab
human
MIF
cancer


Imciromab
mab
mouse
cardiac myosin
cardiac imaging


Imgatuzumab
mab
humanized
EGFR
cancer


Inclacumab
mab
human
selectin P
cardiovascular disease


Indatuximab
mab
chimeric
SDC1
cancer


ravtansine


Indusatumab
mab
human
GUCY2C
cancer


vedotin


Inebilizumab
mab
humanized
CD19
cancer, systemic






sclerosis, multiple sclerosis


Infliximab
mab
chimeric
TNF-α
rheumatoid arthritis,






ankylosing spondylitis,






psoriatic arthritis, psoriasis,






Crohn's disease, ulcerative






colitis


Inolimomab
mab
mouse
CD25 (α chain of IL-
graft versus host disease





2 receptor)


Inotuzumab
mab
humanized
CD22
ALL


ozogamicin


Intetumumab
mab
human
CD51
solid tumors (prostate cancer,






melanoma)


Ipilimumab
mab
human
CD152
melanoma


Iratumumab
mab
human
CD30 (TNFRSF8)
Hodgkin's lymphoma


Isatuximab
mab
chimeric
CD38
cancer


Ixekizumab
mab
humanized
IL 17A
autoimmune diseases


Keliximab
mab
chimeric
CD4
chronic asthma


Labetuzumab
mab
humanized
CEA
colorectal cancer


Lampalizumab
mab
humanized
CFD
geographic atrophy






secondary to age-related






macular degeneration


Lanadelumab
mab
human
kallikrein
angioedema


Landogrozumab
mab
humanized
GDF-8
muscle wasting disorders


Lebrikizumab
mab
humanized
IL-13
asthma


Lemalesomab
mab
mouse
NCA-90 (granulocyte
diagnostic agent





antigen)


Lerdelimumab
mab
human
TGF beta 2
reduction of scarring






after glaucoma surgery


Lexatumumab
mab
human
TRAIL-R2
cancer


Libivirumab
mab
human
hepatitis B surface antigen
hepatitis B


Lifastuzumab
mab
humanized
phosphate-sodium co-
cancer


vedotin


transporter


Ligelizumab
mab
humanized
IGHE
severe asthma and chronic






spontaneous urticaria


Lilotomab
mab
mouse
CD37
cancer


satetraxetan


Lintuzumab
mab
humanized
CD33
cancer


Lirilumab
mab
human
KIR2D
solid and hematological






cancers


Lodelcizumab
mab
humanized
PCSK9
hypercholesterolemia


Lorvotuzumab
mab
humanized
CD56
cancer


mertansine


Lucatumumab
mab
human
CD40
multiple myeloma, non-






Hodgkin's






lymphoma, Hodgkin's






lymphoma


Lulizumab pegol
mab
humanized
CD28
autoimmune diseases


Lumiliximab
mab
chimeric
CD23 (IgE receptor)
chronic lymphocytic






leukemia


Lumretuzumab
mab
humanized
ERBB3 (HER3)
cancer


MABp1
mab
human
IL1A
colorectal cancer


Mapatumumab
mab
human
TRAIL-R1
cancer


Margetuximab
mab
humanized
ch4D5
cancer


Matuzumab
mab
humanized
EGFR
colorectal, lung and stomach






cancer


Mavrilimumab
mab
human
GMCSF receptor α-chain
rheumatoid arthritis


Mepolizumab
mab
humanized
IL-5
asthma and white blood cell






diseases


Metelimumab
mab
human
TGF beta 1
systemic scleroderma


Milatuzumab
mab
humanized
CD74
multiple myeloma and other






hematological malignancies


Mirvetuximab
mab
chimeric
folate receptor alpha
cancer


soravtansine


Mitumomab
mab
mouse
GD3 ganglioside
small cell lung carcinoma


Mogamulizumab
mab
humanized
CCR4
cancer


Motavizumab
mab
humanized
respiratory syncytial virus
respiratory syncytial virus






(prevention)


Moxetumomab
mab
mouse
CD22
cancer


pasudotox


Muromonab-CD3
mab
mouse
CD3
prevention of organ






transplant rejections


Nacolomab
Fab
mouse
C242 antigen
colorectal cancer


tafenatox


Naptumomab
Fab
mouse
5T4
non-small cell lung


estafenatox



carcinoma, renal cell






carcinoma


Narnatumab
mab
human
RON
cancer


Natalizumab
mab
humanized
integrin α4
multiple sclerosis, Crohn's






disease


Nebacumab
mab
human
endotoxin
sepsis


Necitumumab
mab
human
EGFR
non-small cell lung






carcinoma


Nemolizumab
mab
humanized
IL31RA
eczema


Nesvacumab
mab
human
angiopoietin 2
cancer


Nimotuzumab
mab
humanized
EGFR
squamous cell carcinoma,






head and neck






cancer, nasopharyngeal






cancer, glioma


Nivolumab
mab
human
PD-1
cancer


Obiltoxaximab
mab
chimeric

Bacillus anthracis anthrax


Bacillus anthracis spores



Obinutuzumab
mab
humanized
CD20
Chronic lymphatic leukemia


Ocaratuzumab
mab
humanized
CD20
cancer


Ocrelizumab
mab
humanized
CD20
rheumatoid arthritis, lupus






erythematosus etc.


Odulimomab
mab
mouse
LFA-1 (CD11a)
prevention of organ






transplant rejections,






immunological diseases


Ofatumumab
mab
human
CD20
chronic lymphocytic






leukemia etc.


Olaratumab
mab
human
PDGF-R α
cancer


Omalizumab
mab
humanized
IgE Fc region
allergic asthma


Onartuzumab
mab
humanized
human scatter factor
cancer





receptor kinase


Ontuxizumab
mab
chimeric/humanized
TEM1
cancer


Opicinumab
mab
human
LINGO-1
multiple sclerosis


Oportuzumab
scFv
humanized
EpCAM
cancer


monatox


Oregovomab
mab
mouse
CA-125
ovarian cancer


Otelixizumab
mab
chimeric/humanized
CD3
diabetes mellitus type 1


Otlertuzumab
mab
humanized
CD37
cancer


Oxelumab
mab
human
OX-40
asthma


Ozanezumab
mab
humanized
NOGO-A
ALS and multiple sclerosis


Ozoralizumab
mab
humanized
TNF-α
inflammation


Pagibaximab
mab
chimeric
lipoteichoic acid
sepsis (Staphylococcus)


Palivizumab
mab
humanized
F protein of respiratory
respiratory syncytial virus





syncytial virus
(prevention)


Panitumumab
mab
human
EGFR
colorectal cancer


Pankomab
mab
humanized
tumor specific
ovarian cancer





glycosylation of MUC1


Panobacumab
mab
human

Pseudomonas aeruginosa


Pseudomonas








aeruginosa infection



Parsatuzumab
mab
human
EGFL7
cancer


Pascolizumab
mab
humanized
IL-4
asthma


Pasotuxizumab
mab
chimeric/humanized
folate hydrolase
cancer


Pateclizumab
mab
humanized
LTA
TNF


Patritumab
mab
human
ERBB3 (HER3)
cancer


Pembrolizumab
mab
humanized
PDCD1
melanoma and other cancers


Perakizumab
mab
humanized
IL 17A
arthritis


Pertuzumab
mab
humanized
HER2/neu
cancer


Pexelizumab
scFv
humanized
C5
reduction of side effects






of cardiac surgery


Pidilizumab
mab
humanized
PD-1
cancer and infectious






diseases


Pinatuzumab
mab
humanized
CD22
cancer


vedotin


Pintumomab
mab
mouse
adenocarcinoma antigen
adenocarcinoma (imaging)


Placulumab
mab
human
human TNF
pain and inflammatory






diseases


Plozalizumab
mab
humanized
CCR2
diabetic






nephropathy and






arteriovenous graft






patency


Polatuzumab
mab
humanized
CD79B
cancer


vedotin


Ponezumab
mab
humanized
human beta-amyloid
Alzheimer's disease


Priliximab
mab
chimeric
CD4
Crohn's disease, multiple






sclerosis


Pritumumab
mab
human
vimentin
brain cancer


Quilizumab
mab
humanized
IGHE
asthma


Racotumomab
mab
mouse
N-glycolylneuraminic acid
cancer


Radretumab
mab
human
fibronectin extra domain-B
cancer


Rafivirumab
mab
human
rabies virus glycoprotein
rabies (prophylaxis)


Ralpancizumab
mab
humanized
neural apoptosis-regulated
dyslipidemia





proteinase 1


Ramucirumab
mab
human
VEGFR2
solid tumors


Ranibizumab
Fab
humanized
VEGF-A
macular degeneration (wet






form)


Raxibacumab
mab
human
anthrax toxin, protective
anthrax (prophylaxis and





antigen
treatment)


Refanezumab
mab
humanized
myelin-associated
recovery of motor function





glycoprotein
after stroke


Regavirumab
mab
human
cytomegalovirus glycoprotein
cytomegalovirus infection





B


Reslizumab
mab
humanized
IL-5
inflammations of the






airways, skin and






gastrointestinal tract


Rilotumumab
mab
human
HGF
solid tumors


Rinucumab
mab
human
platelet-derived growth
neovascular age-related





factor receptor beta
macular degeneration


Rituximab
mab
chimeric
CD20
lymphomas, leukemias, some






autoimmune disorders


Robatumumab
mab
human
IGF-1 receptor (CD221)
cancer


Romosozumab
mab
humanized
sclerostin
osteoporosis


Rontalizumab
mab
humanized
IFN-α
systemic lupus






erythematosus


Rovelizumab
mab
humanized
CD11, CD18
haemorrhagic shock etc.


Ruplizumab
mab
humanized
CD154 (CD40L)
rheumatic diseases


Sacituzumab
mab
humanized
tumor-associated calcium
cancer


govitecan


signal transducer 2


Samalizumab
mab
humanized
CD200
cancer


Sarilumab
mab
human
IL6
rheumatoid






arthritis, ankylosing






spondylitis


Satumomab
mab
mouse
TAG-72
cancer (diagnosis)


pendetide


Secukinumab
mab
human
IL 17A
uveitis, rheumatoid






arthritis psoriasis


Seribantumab
mab
human
ERBB3 (HER3)
cancer


SGN-CD19A
mab
humanized
CD19
acute lymphoblastic






leukemia and B-cell non-






Hodgkin lymphoma


SGN-CD33A
mab
humanized
CD33
Acute myeloid leukemia


Sibrotuzumab
mab
humanized
FAP
cancer


Sifalimumab
mab
humanized
IFN-α
SLE, dermatomyositis, poly






myositis


Siltuximab
mab
chimeric
IL-6
cancer


Simtuzumab
mab
humanized
LOXL2
fibrosis


Siplizumab
mab
humanized
CD2
psoriasis, graft-versus-host






disease (prevention)


Sirukumab
mab
human
IL-6
rheumatoid arthritis


Sofituzumab
mab
humanized
CA-125
ovarian cancer


vedotin


Solanezumab
mab
humanized
beta amyloid
Alzheimer's disease


Stamulumab
mab
human
myostatin
muscular dystrophy


Sulesomab
Fab′
mouse
NCA-90 (granulocyte
osteomyelitis (imaging)





antigen)


Suvizumab
mab
humanized
HIV-1
viral infections


Tabalumab
mab
human
BAFF
B-cell cancers


Tacatuzumab
mab
humanized
alpha-fetoprotein
cancer


tetraxetan


Tadocizumab
Fab
humanized
integrin αIIbβ3
percutaneous coronary






intervention


Talizumab
mab
humanized
IgE
allergic reaction


Tanezumab
mab
humanized
NGF
pain


Taplitumomab
mab
mouse
CD19
cancer


paptox


Tarextumab
mab
human
Notch receptor
cancer


Tefibazumab
mab
humanized
clumping factor A

Staphylococcus








aureus infection



Tenatumomab
mab
mouse
tenascin C
cancer


Teneliximab
mab
chimeric
CD40
autoimmune diseases and






prevention of organ






transplant rejection


Teplizumab
mab
humanized
CD3
diabetes mellitus type 1


Teprotumumab
mab
human
IGF-1 receptor (CD221)
hematologic tumors


Tetulomab
mab
humanized
CD37
cancer


Tezepelumab
mab
human
TSLP
asthma, atopic dermatitis


Ticilimumab
mab
human
CTLA-4
cancer


(=tremelimumab)


Tigatuzumab
mab
humanized
TRAIL-R2
cancer


Tildrakizumab
mab
humanized
IL23
immunologically mediated






inflammatory disorders


Tocilizumab
mab
humanized
IL-6 receptor
rheumatoid arthritis


(=atlizumab)


Toralizumab
mab
humanized
CD154 (CD40L)
rheumatoid arthritis, lupus






nephritis etc.


Tovetumab
mab
human
CD140a
cancer


Tralokinumab
mab
human
IL-13
asthma etc.


Trastuzumab
mab
humanized
HER2/neu
breast cancer


Trastuzumab
mab
humanized
HER2/neu
breast cancer


emtansine


Tremelimumab
mab
human
CTLA-4
cancer


Trevogrumab
mab
human
growth differentiation
muscle atrophy due to





factor 8
orthopedic disuse






and sarcopenia


Tucotuzumab
mab
humanized
EpCAM
cancer


celmoleukin


Ublituximab
mab
chimeric
MS4A1
cancer


Ulocuplumab
mab
human
CXCR4 (CD184)
hematologic malignancies


Urelumab
mab
human
4-1BB (CD137)
cancer etc.


Urtoxazumab
mab
humanized

Escherichia coli

diarrhea caused by E. coli


Ustekinumab
mab
human
IL-12, IL-23
multiple sclerosis, psoriasis,






psoriatic arthritis


Utomilumab
mab
human
4-1BB (CD137)
cancer


Vandortuzumab
mab
humanized
STEAP1
cancer


vedotin


Vantictumab
mab
human
Frizzled receptor
cancer


Vanucizumab
mab
humanized
angiopoietin 2
cancer


Varlilumab
mab
human
CD27
solid tumors and






hematologic malignancies


Vedolizumab
mab
humanized
integrin α4β7
Crohn's disease, ulcerative






colitis


Veltuzumab
mab
humanized
CD20
non-Hodgkin's lymphoma


Vepalimomab
mab
mouse
AOC3 (VAP-1)
inflammation


Vesencumab
mab
human
NRP1
solid malignancies


Visilizumab
mab
humanized
CD3
Crohn's disease, ulcerative






colitis


Vobarilizumab
mab
humanized
IL6R
inflammatory autoimmune






diseases


Volociximab
mab
chimeric
integrin α5β1
solid tumors


Vorsetuzumab
mab
humanized
CD70
cancer


mafodotin


Votumumab
mab
human
tumor antigen CTAA16.88
colorectal tumors


Zalutumumab
mab
human
EGFR
squamous cell carcinoma of






the head and neck


Zanolimumab
mab
human
CD4
rheumatoid arthritis,






psoriasis, T-cell lymphoma


Zatuximab
mab
chimeric
HER1
cancer


Zolimomab aritox
mab
mouse
CD5
systemic lupus






erythematosus, graft-versus-






host disease









Representative therapeutic uses (e.g., approved indications and proposed indications) for (and targets of) the monoclonal antibodies are set forth in Tables 1 and 2. In some embodiments, the patient (e.g., human) has been diagnosed with a disease or condition such as Crohn's disease, ulcerative colitis or indeterminant colitis, cancer (e.g., neuroblastoma, multiple myeloma, non-small cell lung cancer, Merkel cell cancer, leukemia, colorectal cancer, sarcoma, lymphoma, breast cancer, gastric cancer and melanoma), transplant rejection, hypercholesterolemia, Clostridium difficle infection, sepsis, osteoporosis, multiple sclerosis, anthrax and asthma.


The co-administration of colchicine and/or hydroxychloroquine with a therapeutic monoclonal antibody may be particularly advantageous in chronically administered monoclonal antibodies, since duration of the treatment with the monoclonal antibodies contributes to the development of anti-drug antibodies. Examples of monoclonal antibodies with long duration of use include those that are indicated for the treatment of autoimmune diseases. In particular, inhibitors of tumor necrosis factor alpha (TNF-α) that are used to treat the inflammatory bowel diseases (ulcerative colitis and Crohn's disease) are commonly prescribed together with thiopurines to prevent antibody formation. These TNF-α inhibitors would include, but are not limited to: adalimumab, certolizumab, golimumab, infliximab and ozoralizumab. Yet other monoclonal antibodies used to treat inflammatory bowel diseases that may be particularly suited for use the present invention include inhibitors of integrin a (e.g., abrilumab, etaracizumab, etrolizumab, natalizumab, vedolizumab, volociximab). Cancer is yet another disease that may entail prolonged treatment with a monoclonal antibody. For example, volociximab is an inhibitor of integrin-α that is used for the treatment of solid tumors. Thus, anti-cancer monoclonal antibodies may also be useful in practice of the present invention.


The terms “co-administering” or “co-administration” as used herein embrace administration of the therapeutically effective amount of the monoclonal antibody and the effective amount of hydroxychloroquine and/or colchicine simultaneously, either in the same or different dosage forms, or at different times, provided that they are made during the treatment “period” which for purposes of the present invention, includes the time while the monoclonal antibody is still present in the blood serum. That is, the monoclonal antibody and hydroxychloroquine and/or colchicine can be administered together or separately, for example, at different times and in different formulations and/or via different routes of administration. In some embodiments, hydroxychloroquine and/or colchicine may be administered to the patient can be prior to initiation of the monoclonal antibody therapy, e.g., to build up levels of hydroxychloroquine in the system to prevent anti-drug antibody formation. In some embodiments, hydroxychloroquine and/or colchicine may be initiated at the time of the monoclonal antibody therapy and may continue for the duration of therapy. In various embodiments, co-administering hydroxychloroquine and/or colchicine to the patient can be after administering the monoclonal antibody.


Suitable dosages for the active compounds such as a monoclonal antibody, colchicine and/or hydroxychloroquine can be those dosages presently used in connection with approved indications. Advantages of the present invention, however, are that these dosages can be lowered and/or administered less frequently due to the combined action of the two agents.


Therapeutically effective amounts of monoclonal antibodies depend upon many factors, including for example, the nature and severity of the disease or condition, the age and general health and weight of the patient. Generally, therapeutically effective dosage amounts are known in the art.


Hydroxychloroquine and colchicine may be administered as a free base or in the form of a pharmaceutically acceptable hydrate, solvate or salt. All such forms are embraced by the terms “hydroxychloroquine” and “colchicine”. Hydroxychloroquine is advantageously administered in the form of a sulfate salt. In some embodiments, administration of hydroxychloroquine and/or colchicine to the patent can include daily administration, or every other day, to the patient during monoclonal antibody therapy of the patient. Administering hydroxychloroquine and/or colchicine can begin days or weeks before beginning monoclonal antibody therapy, or can being contemporaneous with initiating monoclonal antibody therapy, or after such therapy has begun. Hydroxychloroquine and/or colchicine may also be administered every third day, every fourth day, or weekly to the patient.


An “effective amount” of the hydroxychloroquine embraces amounts that result in a clearance time of the monoclonal antibody from the circulation or blood serum of the patient that is increased relative to the same regimen of treatment with the monoclonal antibody but without the co-administration of colchicine and/or hydroxychloroquine. In some embodiments, the daily dose of hydroxychloroquine ranges between about 5 mg and about 800 mg, between about 5 mg and about 600 mg, between about 25 mg and about 600 mg, or between about 100 mg and about 400 mg. In some embodiments, dosing of hydroxychloroquine may include an initial dosing following by a maintenance dosing schedule. Thus, for example, an initial dose (in the form of the sulfate salt) may range from about 25 to about 600 mg (19.4 to 465 mg base), taken orally once daily, or in some other embodiments an initial loading of 800 mg. The initial dose may be administered from one to about twelve weeks. A maintenance dose (in the form of the sulfate salt) may range from about 5 to about 400 mg (3.9 mg to 310 mg base) taken orally once daily. Hydroxychloroquine—sulfate salt may be commercially available (PLAQUENIL) in the form of 200 mg tablets.


In general, the daily dose of colchicine may range from about 0.05 mg to about 5 mg, and in some embodiments from about 0.07 mg to about 3.5 mg, and in some other embodiments, from about 0.08 mg to about 3 mg, and in yet other embodiments from about 0.1 mg to about 2.4 mg.


Those skilled in the art appreciate that the dosage regimen of hydroxychloroquine may be adjusted, depending upon the needs of the patient. For example, the dose may need to be reduced, at least temporarily, if the patient develops any adverse side effects. Then after 5 to 10 days the dose may gradually be increased to a recommended final dose. Hydroxychloroquine is advantageously administered with a solid or liquid meal (e.g., milk).


In terms of duration of a therapy period, treatment with the monoclonal antibody to the patient may include administering the patient in intervals of about one week, about two weeks, about three weeks, about four weeks, about five weeks, about six weeks, about seven weeks, about eight weeks, about nine weeks, about 10 weeks, about 11 weeks, or about 12 weeks. That is, the interval can be about one week to about 12 weeks, including each of the other time intervals disclosed herein. In view of the advantages of the present invention, however, not only can the dosage amounts be decreased, alternatively or in conjunction therewith, the monoclonal antibody can be administered less frequently in comparison to administering the monoclonal antibody to a patient not being administered hydroxychloroquine and/or colchicine. For example, where a monoclonal antibody is administered such as infused in eight week intervals, the combination therapy of the present invention may extend the administration of the monoclonal antibody to 10 week or 12 week intervals.


Co-administration also entails administration of each agent in accordance via routes known to be effective and safe. For example, the present methods may include administering hydroxychloroquine and/or colchicine orally, and in the case of colchicine, orally or parenterally (e.g., intravenously). The present invention may include administering the monoclonal antibody to the patient subcutaneously such as intravenously. By way of illustration, infliximab can be administered intravenously and hydroxychloroquine and/or colchicine can be administered orally. As another example, adalimumab can be administered subcutaneously and hydroxychloroquine and/or colchicine can be administered orally. By way of additional illustration, infliximab can be administered intravenously and colchicine can be administered intravenously. As another example, adalimumab can be administered subcutaneously and colchicine can be administered intravenously.


Therapeutic combinations of the present invention include a therapeutically effective amount of a monoclonal antibody, and an effective amount of colchicine and/or hydroxychloroquine, or a combination thereof. These agents may be formulated in the same or different dosage forms.


Monoclonal antibodies are typically formulated for parenteral (e.g., intravenous, intraperitoneal, infusion, intraarterial, intramuscular, subcutaneous) administration. Colchicine may also be formulated for parenteral administration, which may be advantageous in embodiments wherein the monoclonal antibody is being used as an anti-cancer agent. Pharmaceutically acceptable carriers or vehicles include nontoxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives (e.g., octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens, such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counterions such as sodium; metal complexes (e.g., Zn-protein complexes); and non-ionic surfactants such as TWEEN or polyethylene glycol (PEG). More particularly, preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media, such as 0.01-0.1M and preferably 0.05M phosphate buffer or 0.8% saline. Other common parenteral carriers or vehicles include sodium phosphate solutions, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, and fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like.


The composition should be sterile and fluid for purposes of ease of syringability. Sterile injectable solutions can be prepared by incorporating the monoclonal antibody and the vehicle, in the required amount followed by filtered sterilization. Generally, dispersions are prepared by incorporating the monoclonal antibody into a sterile vehicle including a basic dispersion medium. In the case of sterile powders for the preparation of sterile injectable solutions, one method of preparation is vacuum drying and freeze-drying, which yields a powder of the monoclonal antibody from a previously sterile-filtered solution thereof. The preparations for injections are processed, filled into containers such as ampoules, bags, bottles, syringes or vials, and sealed under aseptic conditions according to methods known in the art.


As may be appropriate, other formulation types and routes of administration (e.g., topical, transdermal, oral, rectal, pulmonary) may be appropriate, depending for example on the monoclonal antibody and the indication being treated.


In some embodiments, hydroxychloroquine and/or colchicine is administered orally, optionally in combination with one of more conventional pharmaceutically acceptable carriers and/or excipients. Oral formulations containing hydroxychloroquine and/or colchicine may include tablets, capsules, buccal forms, troches, lozenges and oral liquids such as suspensions and solutions. Capsules can contain mixtures of active compound(s) with inert filler(s) and/or diluent(s) such as the pharmaceutically acceptable starches (e.g., corn, potato or tapioca starch), sugars, artificial sweetening agents, powdered celluloses), flours, gelatins, gums, and the like. Tablet formulations can be made by conventional compression, wet granulation or dry granulation methods and utilize pharmaceutically acceptable diluents, binding agents, lubricants, disintegrants, surface modifying agents (including surfactants), suspending or stabilizing agents, including magnesium stearate, stearic acid, sodium lauryl sulfate, talc, sugars, lactose, dextrin, starch, gelatin, cellulose, methyl cellulose, microcrystalline cellulose, sodium carboxymethyl cellulose, carboxymethyl cellulose calcium, polyvinylpyrrolidine, alginic acid, acacia gum, xanthan gum, sodium citrate, complex silicates, calcium carbonate, glycine, sucrose, sorbitol, dicalcium phosphate, calcium sulfate, lactose, kaolin, mannitol, sodium chloride, low melting waxes, and ion exchange resins. Preferred surface modifying agents include nonionic and anionic surface modifying agents. Representative examples of surface modifying agents include poloxamer 188, benzalkonium chloride, calcium stearate, cetostearl alcohol, cetomacrogel emulsifying wax, sorbitan esters, colloidal silicon dioxide, phosphates, sodium dodecylsulfate, magnesium aluminum silicate, and triethanolamine. Oral formulations herein can utilize standard delay or time release formulations to alter the absorption of the hydroxychloroquine.


In some embodiments, hydroxychloroquine may be formulated in the form of a tablet, along with pharmaceutically acceptable carriers and excipients, including dibasic calcium phosphate, hydroxypropyl methylcellulose, magnesium stearate, polyethylene glycol 400, Polysorbate 80, starch and titanium dioxide. In some embodiments, colchicine is administered in the form of a tablet.


Pharmaceutically acceptable liquid carriers include water, organic solvents, mixtures of both, and pharmaceutically acceptable oils or fats. The compositions may also contain one or more pharmaceutically acceptable excipients or additives such as solubilizers, emulsifiers, buffers, preservatives, sweeteners, flavoring agents, suspending agents, thickening agents, colors, viscosity regulators, stabilizers, and osmo-regulators.


Kits of the present invention include the therapeutic combination, which in turn includes a therapeutically effective amount of a monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof. The kit may include both agents in a single dosage form or in separate dosage forms, in which case the respective dosage forms may be disposed in separate containers in the kit. The kit may further include a label or insert that includes printed instructions for using the therapeutic combination to practice the methods described herein. The instructions may be those customarily used in commercial packages of therapeutic products and may contain information about indications, usage, dosage, administration, contraindications and/or warnings concerning use of the products, etc.


Suitable containers include, for example, bottles, vials, syringes, blister pack, and the like. The container can be formed from a variety of materials such as glass or plastic. The container can hold a monoclonal antibody and the like or a formulation thereof which is effective, for treating the condition and may have a sterile access port (e.g., the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The kit may further include another container including a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. A kit can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. Thus, in some embodiments, a kit may include a first container with a monoclonal antibody contained therein, a second container with a hydroxychloroquine and/or colchicine contained therein, and optionally, a third container including a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.


In some embodiments, the kits are customized for the delivery of solid oral forms of hydroxychloroquine and/or colchicine, such as by tablets or capsules. Such a kit can include a number of unit dosages, such as a card having the dosages oriented in the order of their intended use. An example of such a kit is a “blister pack”. Blister packs are well known in the package industry and are widely used for packaging pharmaceutical unit dosage forms. If desired, a memory aid can be provided, for example, in the form of numbers, letters, or other markings or with a calendar insert, designating the days in the treatment schedule in which the dosages can be administered. In other embodiments, the kits are customized for the delivery of a parenteral delivery of colchicine, which may be included in the same dosage formulation as the monoclonal antibody, or a different dosage formulation.


In some embodiments, the kit may include a container for containing the separate pharmaceutical compositions such as a divided bottle or a divided foil packet; however, the separate pharmaceutical compositions can be contained with a single, undivided container. Typically, the kit includes descriptions for the co-administration of the separate compositions. Kits of the present invention may be particularly advantageous when the separate compositions are administered in different dosage forms (e.g., hydroxychloroquine and/or orally and a monoclonal antibody parenterally), and/or are administered at different dosage intervals, and/or when titration of the individual components of the therapeutic combination is desired by the prescribing physician.


The present invention may also have utility in diagnostic applications, where the monoclonal antibody is being used to detect the presence or severity of a disease or other pathological condition. Thus, further aspects of the present invention may include a method for enhancing the efficacy of monoclonal antibody diagnosis, which entails co-administering a diagnostically effective amount of a monoclonal antibody, or a functional fragment thereof, which is optionally labeled (e.g., with a radio-label or a fluorescent label), and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof. A related aspect is directed to a method of increasing the time a diagnostic monoclonal antibody remains in the circulation of a patient, which entails co-administering an effective amount of the diagnostic monoclonal antibody which is optionally labeled, or a functional fragment thereof, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof, wherein the time the monoclonal antibody remains in the circulation (e.g., blood serum) of the patient is increased relative to the same regimen of administration of the diagnostic monoclonal antibody but without the co-administration of effective amount of the colchicine and/or hydroxychloroquine. Yet another aspect of the present invention is directed to a diagnostic combination, which includes a diagnostically effective amount of an optionally labeled monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof. A further aspect of the present invention is directed to a kit, which includes a diagnostic combination, which includes a diagnostically effective amount of an optionally labeled monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof. The kit may include both agents in a single dosage form or in separate dosage forms, in which case the respective dosage forms may be disposed in separate containers in the kit. The kit may further include printed instructions for using the diagnostic combination to practice the methods described herein. Diagnostic labels and amounts of antibodies for use in diagnostic methods are known in the art.


All publications cited in the specification, including patent publications and non-patent publications, are indicative of the level of skill of those skilled in the art to which this invention pertains. All these publications are herein incorporated by reference to the same extent as if each individual publication were specifically and individually indicated as being incorporated by reference.


Although the invention described herein has been described with reference to particular embodiments, it is to be understood that these embodiments are merely illustrative of the principle and applications described herein. It is therefore to be understood that numerous modifications may be made to the illustrative embodiments and that other arrangements may be devised without departing from the spirit and scope of the various embodiments described herein as defined by the amended claims.

Claims
  • 1. A method for enhancing the efficacy of monoclonal antibody therapy, which entails co-administering a therapeutic monoclonal antibody, or a functional fragment thereof, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof.
  • 2. A method of increasing time that a monoclonal antibody remains in the circulation of a patient, which entails co-administering a therapeutic monoclonal antibody, or a functional fragment of the monoclonal antibody, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof, to a patient in need thereof, wherein the time the monoclonal antibody remains in the circulation of the patient is increased relative to the same regimen of administration of the monoclonal antibody but without the effective amount co-administration of colchicine and/or hydroxychloroquine.
  • 3. The method of claim 1, wherein the monoclonal antibody is administered parenterally.
  • 4. The method of claim 3, wherein the monoclonal antibody is administered intravenously.
  • 5. The method of claim 3, wherein the monoclonal antibody is administered subcutaneously.
  • 6. The method of claim 1, wherein the monoclonal antibody is a human monoclonal antibody.
  • 7. The method of claim 1, wherein the monoclonal antibody is a humanized monoclonal antibody.
  • 8. The method of claim 1, wherein the monoclonal antibody is a chimeric monoclonal antibody.
  • 9. The method of claim 1, wherein the monoclonal antibody is a murine monoclonal antibody.
  • 10. The method of claim 1, wherein the monoclonal antibody is conjugated.
  • 11. The method of claim 10, wherein the monoclonal antibody is conjugated to a drug.
  • 12. The method of claim 1, wherein the monoclonal antibody inhibits tissue necrosis factor—alpha (TNF-α).
  • 13. The method of claim 12, wherein the monoclonal antibody is selected from the group consisting of adalimumab, certolizumab pegol, golimumab, and infliximab.
  • 14. The method of claim 13, wherein the monoclonal antibody is infliximab.
  • 15. The method of claim 1, wherein the hydroxychloroquine and/or colchicine is administered orally.
  • 16. The method of claim 1, wherein the hydroxychloroquine is administered in the form of a sulfate salt.
  • 17. The method of claim 16, wherein the hydroxychloroquine is administered in an amount of about 5 mg to about 800 mg.
  • 18. The method of claim 1, wherein the monoclonal antibody is an inhibitor of integrin-α.
  • 19. The method of claim 1, wherein colchicine is administered parenterally.
  • 20. A therapeutic combination, comprising a therapeutically effective amount of a monoclonal antibody, or a functional fragment thereof, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof.
  • 21. A kit, comprising a therapeutic combination comprising a therapeutically effective amount of a monoclonal antibody, or a functional fragment thereof, and an effective amount of colchicine or hydroxychloroquine, or a combination thereof.
  • 22. The kit of claim 21, comprising a first container comprising the monoclonal antibody in a formulation suitable for parental administration, and a second container comprising the hydroxychloroquine, colchicine or a combination thereof, in a dosage form suitable for oral administration.
  • 23. The kit of claim 21, comprising a first container comprising the monoclonal antibody in a formulation suitable for parental administration, and a second container comprising the colchicine in a dosage form suitable for parenteral administration.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation-in-part of U.S. patent application Ser. No. 14/947,193, filed Nov. 20, 2015, which claims the benefit of U.S. Provisional Application No. 62/082,692, filed Nov. 21, 2014. The disclosures of each of these are incorporated herein by reference in their entireties for all purposes.

Provisional Applications (1)
Number Date Country
62082682 Nov 2014 US
Continuation in Parts (1)
Number Date Country
Parent 14947193 Nov 2015 US
Child 15605212 US