Compositions of bosutinib with anti-HER 1, anti-HER 2, or anti-HER 4 antibodies and methods of use thereof

文档序号:913524 发布日期:2021-02-26 浏览:8次 中文

阅读说明:本技术 波齐替尼与抗-her1抗体、抗-her2抗体或抗-her4抗体的组合物及其使用方法 (Compositions of bosutinib with anti-HER 1, anti-HER 2, or anti-HER 4 antibodies and methods of use thereof ) 是由 古鲁·雷迪 张顺英 卞周允 于 2019-06-25 设计创作,主要内容包括:本发明提供了一种波齐替尼和抗-HER1抗体、抗-HER2抗体或抗-HER4抗体的组合物(可选地与其他药物一起使用),以及所述组合物在治疗癌症中的应用。(The invention provides a composition of bosutinib and an anti-HER 1 antibody, an anti-HER 2 antibody or an anti-HER 4 antibody (optionally together with other drugs), and the use of the composition in the treatment of cancer.)

1. A method of treating cancer in a subject, comprising administering to the subject a therapeutically effective amount of bosutinib and an anti-HER 1 antibody, an anti-HER 2 antibody, or an anti-HER 4 antibody;

wherein the cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER 4;

the cancer is non-small cell lung cancer, breast cancer, colon cancer, stomach cancer, pancreatic cancer, prostate cancer, myeloma, head cancer, neck cancer, ovarian cancer, esophageal cancer or metastatic cell cancer.

2. The method of claim 1, further comprising administering at least one of paclitaxel, cisplatin, 5-fluorouracil, vinorelbine, and cetuximab, or any pharmaceutical combination thereof.

3. The method of claim 1 or 2, wherein the antibody is an anti-HER 2 antibody selected from trastuzumab, cetuximab, or any antigen-binding fragment thereof.

4. The method of any one of claims 1-3, wherein the cancer is breast cancer.

5. The method of claim 4, wherein the breast cancer is selected from

(i) Estrogen receptor negative breast cancer, HER1 and/or HER2 overexpression;

(ii) estrogen receptor and progesterone receptor double positive breast cancer, HER2, is expressed but not overexpressed;

(iii) trastuzumab-resistant breast cancer, HER2 overexpression;

(iv) PR, HER2 and estrogen receptor negative breast cancer, HER1 are overexpressed.

6. The method of claim 5, wherein the breast cancer is metastatic breast cancer.

7. The method of claim 6, further comprising the steps of:

collecting the subject's breast cancer cells;

the breast cancer cells are evaluated to confirm overexpression of HER2, overexpression of a HER2 mutant, amplification of HER2, or amplification of a HER2 mutant.

8. The method of claim 7, wherein said assessing comprises applying Immunohistochemistry (IHC) and Fluorescence In Situ Hybridization (FISH) techniques.

9. The method of claim 8, wherein the IHC is IHC3+ or IHC2 +.

10. A method of treating breast cancer in a subject in need thereof, wherein the breast cancer is associated with overexpression or amplification of HER2, or overexpression or amplification of a HER2 mutant, the method comprising the steps of:

a) within a 21 day (± 3 days) cycle

i) Administering 11.5-5.5 mg/kg of T-DM in a single dose;

ii) 0.5-50 mg/d of bosutinib is administered daily; and

iii) optionally discontinuing the drug for a period of 21 days;

b) optionally repeating the cycle, or adjusting the cycle.

11. The method of claim 10, wherein the 21-day cycle comprises a 2-week dosing period and a 1-week drug holiday.

12. The method of claim 11, wherein the 21-day cycle is selected from

After two weeks of administration, take out the drug for one week;

one week after dosing, followed by one more week after dosing; alternatively, the first and second electrodes may be,

one week after withdrawal, two weeks after withdrawal.

13. The method of any one of claims 10 to 12, wherein the administration of T-DM1 is intravenous infusion.

14. The method of claim 13, wherein the standard dose of T-DM1 is 3.6 mg/kg.

15. The method of any one of claims 10-14, wherein the administration of the bociclib is oral.

16. The method of claim 15, wherein the oral dose is selected from 6, 8,10, 12, 16 or 24mg once daily.

17. A method of ameliorating the occurrence of an adverse event in a subject undergoing treatment for breast cancer associated with overexpression or amplification of HER2 or a mutant of HER2, the method comprising the steps of:

a) within a 21 day (± 3 days) cycle

I) single dose administration of T-DM 1;

ii) administering a daily dose of bosutinib; and

b) optionally the cycle is repeated for a number of times,

wherein the adverse event is selected from cardiac toxicity, hematologic toxicity, diarrhea, rash, mucositis, fatigue, electrolyte abnormalities, or hepatotoxicity.

18. A composition for treating breast cancer in a subject, wherein the breast cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER4, the composition comprising a therapeutically effective amount of bosutinib and T-DM1, wherein the bosutinib is administered orally and T-DM1 is administered intravenously.

19. The composition of claim 18, further comprising a therapeutically effective amount of paclitaxel administered by Intravenous (IV) infusion.

20. A method of treating gastric cancer in a subject, the method comprising administering to the subject a therapeutically effective amount of bosutinib and an anti-HER 1 antibody, an anti-HER 2 antibody, or an anti-HER 4 antibody;

wherein, gastric cancer is related to the overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2 or a mutant of HER 4.

21. The method of claim 20, further comprising administering at least one of paclitaxel, cisplatin, 5-fluorouracil, vinorelbine, and cetuximab, or any pharmaceutical combination thereof.

22. The method of claim 20 or 21, wherein the antibody is an anti-HER 2 antibody selected from trastuzumab, cetuximab, or any antigen-binding fragment.

23. The method of any one of claims 20 to 22, further comprising the preparation step of:

collecting the gastric cancer cells of the subject;

gastric cancer cells were evaluated to confirm overexpression of HER2, overexpression of a mutant of HER2, amplification of HER2 gene, or amplification of a mutant of HER2 gene.

24. The method of any one of claims 20 to 23, wherein said assessing comprises using Immunohistochemistry (IHC) and Fluorescence In Situ Hybridization (FISH) techniques.

25. The method of claim 24, wherein the IHC is IHC3+ or IHC2 +.

26. The method of any one of claims 20-25, wherein the gastric cancer has been treated with chemotherapy.

27. A method of treating gastric cancer in a subject in need thereof, wherein the gastric cancer is associated with overexpression or amplification of HER2 or overexpression or amplification of a HER2 mutant, the method comprising the steps of:

a) within a 21 day (± 3 days) cycle

I) single dose administration of 6-8 mg/kg trastuzumab;

ii) a single dose of 105 to 175mg/m paclitaxel2

Iii) administering the bosutinib 4-16 mg/d daily;

b) the above cycle is optionally repeated.

28. The method of claim 27, wherein trastuzumab was administered by Intravenous (IV) infusion on day 1.

29. The method of claim 27 or 28, wherein the standard dose of trastuzumab is at a payload of 8mg/kg followed by a 6mg/kg infusion.

30. The method of any one of claims 27-29, wherein the paclitaxel is administered by Intravenous (IV) infusion on day 1.

31. The method of claim 30, wherein the standard dose of paclitaxel is 175mg/m2And (4) infusion.

32. The method of any one of claims 27-31, wherein bosutinib is administered orally for 14 ± 3 days.

33. The method of claim 32, wherein the oral dose is selected from 4, 6, 8,10, 12, or 16mg once daily.

34. The method of claim 33, wherein the break is 7 days after 14 days of the administration of the bosutinib.

35. A method of ameliorating the occurrence of an adverse event in a subject undergoing gastric cancer treatment associated with overexpression or amplification of HER2 or a mutant of HER2, the method comprising the steps of:

a) within a 21 day (± 3 days) cycle

i) T-DM1 administered in a single dose;

ii) administering a daily dose of bosutinib; and

b) optionally repeating said cycle;

wherein the adverse event is selected from cardiac toxicity, hematologic toxicity, diarrhea, rash, mucositis, fatigue, electrolyte abnormalities, or hepatotoxicity.

36. A composition for treating gastric cancer in a subject, wherein the gastric cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER4, the composition comprising a therapeutically effective amount of bosutinib and T-DM1, wherein the bosutinib is administered orally and T-DM1 is administered Intravenously (IV).

37. The composition of claim 36, further comprising a therapeutically effective amount of paclitaxel administered by Intravenous (IV) infusion.

Technical Field

The present disclosure relates generally to a method of treating cancer by administering to a subject a pharmaceutical composition comprising a combination of an anti-HER 1 antibody, an anti-HER 2 antibody, or an anti-HER 4 antibody and poetinib, optionally further comprising a combination of other anti-cancer drugs.

Background

The Epidermal Growth Factor Receptor (EGFR) family is known to include 4 members, namely HER1/ErbB1 (commonly referred to as "EGFR"), HER2/ErbB2, HER3/ErbB3, and HER4/ErbB 4. EGFRs play an important role in normal cellular regulation through intracellular signal transduction, and these proteins regulate cell growth, apoptosis, migration, adhesion, and differentiation. The aberrant overexpression or mutation of EGFRs in most solid tumor cells, hyperactivation of these receptors triggers a complex, multi-layered, interrelated signaling pathway network including the downstream mitogen-activated protein kinase (MARK), phosphatidylinositol-3-kinase/protein kinase (PI3K/AKT), and Janus kinase/signal transducer pathways, as well as the transcriptional activator (JAK/STAT) pathway, promoting cancer growth, differentiation, angiogenesis, metastasis, and drug resistance. For example, HER2 is overexpressed in approximately 20% to 25% of breast cancers and serves as a prognostic marker for breast cancer. HER2 positive breast cancer is more clinically aggressive and invasive than the HER2 negative subtype, and is associated with increased tumor growth rate, early systemic metastasis and poor prognosis. Therefore, blocking epidermal growth factor receptor-mediated tumor cell signaling pathway is an ideal target for anti-tumor effect.

Anticancer drugs targeting EGFRs fall into two categories: monoclonal antibodies targeting the extracellular domain and small molecule drugs targeting intracellular tyrosine kinases. The monoclonal antibody is selectively combined with an epidermal growth factor receptor, and has the advantages of good drug effect and small side effect. However, monoclonal antibodies have the disadvantage of being expensive and have to be administered by injection. Meanwhile, small molecule drugs targeting tyrosine kinases are relatively inexpensive and orally available, and they have good drug efficacy by selectively interacting with receptor subtypes (such as EGFR, HER2, HER3, or HER4) or interacting with multiple receptor subtypes simultaneously.

Trastuzumab antibody-drug conjugate (T-DM1) is an example of an antibody-drug conjugate targeting HER2, in which the antibody (humanized anti-HER 2 lgG1, trastuzumab) is covalently linked to the microtubule inhibiting drug DM1, a maytansine derivative, via a stable thioether linker MCC [ i.e. 4- (N-maleimidomethyl) cyclohexane-1-carboxylate ]. "maytansinoid conjugate (Emtansine)" refers to the MCC-DM1 complex. T-DM1 is considered to be a single agent in HER2 positive metastatic breast cancer patients who had received trastuzumab and paclitaxel either as monotherapy or in combination. The structural formula of the trastuzumab-DM 1 conjugate (T-DM1) is shown as follows.

Bosutinib (HM781-36B) is a novel oral irreversible pan-HER inhibitor described in U.S. patent No. 8,188,102, which is incorporated herein by reference in its entirety. Bosutinib is a quinazoline-based tyrosinase kinase inhibitor (structure shown below) that irreversibly blocks signaling through the tyrosinase kinase receptor EGFR family, including HER1(EGFR), HER2, and HER4 wild-type receptors, as well as receptors with activating mutations. And then inhibiting the proliferation of tumor cells that overexpress the above receptors. The administration of bosutinib inhibits the proliferation of tumor cells that overexpress the above receptors. The chemical formula of poqitinib is as follows: 1- [4- [4- (3, 4-dichloro-2-fluorophenylamino) -7-methoxyquinazolin-6-oxy ] -piperidin-1-yl ] prop-2-en-1-one.

In recent years, it has been reported that the expression of drug resistance in EGFR-targeted therapy shortens the duration of therapeutic effect of the drug. Non-small cell lung cancer (NSCLC) patients with EGFR-activating mutations who were treated with gefitinib or erlotinib were reported to develop resistance to the drug after about 8 to 16 months of treatment, and about 60% of patients were observed to develop resistance due to EGFR T790M mutation (helenaa. yu et al, cli. cancer res.19(8),2240,2013). Furthermore, of HER2 positive metastatic breast Cancer patients treated with the antibody drug trastuzumab, 66% to 88% of patients are known to exhibit primary or acquired resistance due to various mechanisms (Alice Chung et al, cli. Breast Cancer 13(4),223,2013). Based on this, although EGFR-targeted treatment of HER 2-overexpressed solid cancers has a significant effect, the therapeutic effect of EGFR-targeted therapeutics cannot be maintained for a long period of time due to the generation of primary and secondary resistance, and thus their development is limited.

Gastric cancer is the fifth most common cancer in the world and is also the third leading cause of cancer-related death. Chemotherapy is the fundamental therapy for most patients with metastatic or recurrent gastric cancer. Although there is currently no generally accepted chemotherapeutic regimen, the use of fluoropyrimidines in combination with platinum drugs is most commonly used. Some physicians have added docetaxel or epirubicin to this double combination. However, triple therapy is not commonly used due to toxicity concerns.

Therefore, there is a strong need for an effective treatment to improve the efficacy and overcome drug resistance in the treatment of EGFR overexpression or mutation, particularly solid cancers of HER 2. The present invention satisfies this need.

Disclosure of Invention

In one aspect, the present disclosure provides a method of treating cancer in a subject, the method comprising administering to the subject a therapeutically effective amount of bosutinib and an anti-HER 1 antibody, an anti-HER 2 antibody, or an anti-HER 4 antibody, antibody conjugate, or fragment thereof, wherein the cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER 4. Suitably, the anti-HER 2 antibody is trastuzumab or a drug conjugate thereof, such as enrmetuzumab (T-DM 1). The method may further comprise administering at least one of paclitaxel, cisplatin, 5-fluorouracil, vinorelbine, cetuximab, or any combination thereof. The method is suitable for non-small cell lung cancer, breast cancer, gastric cancer, colon cancer, pancreatic cancer, prostatic cancer, myeloma, head and neck cancer, ovarian cancer, esophageal cancer, metastatic cell cancer and other cancers. These cancers may be primary or secondary. Suitably, the method of treating cancer is directed to a subject having breast cancer, lung cancer or gastric cancer. Suitably, the breast cancer comprises metastatic breast cancer. Suitably, the method of treating cancer is directed to a subject having gastric cancer, preferably a subject having HER2 positive gastric cancer, more preferably a HER2 positive gastric cancer subject who has been treated with one or more chemotherapeutic drugs.

In another aspect, the present disclosure provides a method of treating breast cancer in a subject, wherein the breast cancer is associated with overexpression or amplification of HER1, HER2, HER4, or a HER2 mutant, wherein the method comprises a) a step within a 21 day (± 3 days) cycle, i) single dose administration of a standard dose of an anti-HER 2 antibody or conjugate thereof, e.g., T-DM 1; and ii) administering a daily dose of bosutinib; and b) optionally repeating said cycle. Suitably, T-DM1 is administered by Intravenous (IV) infusion. Suitably, the standard dose of T-DM1 is 0.5-10 mg/kg. Suitably, the dose of T-DM1 is 3.6 mg/kg. Suitably, the administration of the brigatinib is oral. The oral dose of the brigatinib is 0.5-50 mg/d.

In another aspect, the present disclosure provides a method of ameliorating the occurrence of an adverse event in a subject undergoing treatment for breast cancer associated with overexpression or amplification of HER2 or a HER2 mutant, the method comprising a) within a 21 day (± 3 days) cycle, i) single dose administration of T-DM 1; and ii) administering a daily dose of bosutinib; and b) optionally repeating said cycle.

Another aspect of the present disclosure provides a composition for treating cancer in a subject, wherein the cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER4, and the composition comprises a therapeutically effective amount of bosutinib and T-DM1, wherein bosutinib is administered orally and T-DM1 is administered by intravenous infusion.

The advances and advantages of the present disclosure will become apparent to those of ordinary skill in the art upon reading the following detailed description. While the methods and pharmaceutical compositions for treating cancer are susceptible of various forms of embodiment, the following description includes specific embodiments, with the understanding that these embodiments are illustrative, and are not intended to limit the invention thereto.

Drawings

Figures 1a and 1b show median progression-free survival curves (1a) and median overall survival curves (1b) for patients with combined phase I and phase II;

fig. 2a and 2 b: figure 2a shows a waterfall plot (2a) of the optimal percent change in stage I and II bound target lesion tumor diameter; FIG. 2b shows only the waterfall plot of phase II;

fig. 3 shows the cumulative bar graph of stage ii progression-free survival (Swimmerplot) in example 3, duration of treatment: [ (according to the event occurrence or date of last dose) — date of first dose +1 ]/(365.25/52). The study end date was used if the last dosing date was not collected. The best overall response rate (BOR) is shown on the right side of each study duration bar. []: and (4) determining the BOR. If not specified, the BOR is the same as the determined BOR.

Detailed Description

Definition of

As disclosed herein, the present disclosure provides a number of range values. It is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range of values is specifically disclosed. Each smaller range between any stated value or intervening value in a stated range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included or excluded in the range, and smaller ranges in which only one, neither or both limits are included are also within the scope of the invention, but are limited to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.

As used herein, the term "about" generally refers to plus or minus 10% of the number. For example, "about 10%" may mean a range of 9-11%, and "about 20" may mean 18-22. Other meanings of "about" may be apparent from the context, such as rounding off, so for example "about 1" may also mean 0.5 to 1.4. As used herein, the term "and/or" includes any and all combinations of one or more of the associated listed items. The expression "at least one" modifies an entire list of elements as opposed to modifying individual elements in the list in front of the list. The terms "day", "daily", and the like, when referring to a dosing regimen, refer to the time from the beginning of midnight to the end of the next midnight within a calendar day.

The term "treatment" or "diagnostically treating" and any derivatives thereof, as used herein, refers to a therapeutic treatment. For a particular condition, treatment refers to: (1) ameliorating or preventing a disorder of one or more biological manifestations, (2) interfering with (a) one or more points in a biological cascade leading to the disease or leading to the disease, or (b) one or more biological manifestations of the disease, (3) alleviating one or more symptoms, effects, or side effects associated with the disease or treatment thereof, or (4) slowing the progression of a condition or slowing one or more biological manifestations of a condition. Therefore, prophylactic treatment is also under consideration. The skilled person will appreciate that "prevention" is not an absolute term. In medicine, "prevention" is to be understood as prophylactic administration to substantially reduce the likelihood or severity of, or delay the onset of, a disease or biological manifestation thereof. For example, prophylactic treatment is appropriate when the subject is considered to be at high risk of developing cancer, such as when the subject has a strong family history of cancer or when the subject has been exposed to a carcinogen.

As used herein, the term "therapeutically effective amount" refers to an amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for example, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" refers to any dose that is capable of treating, curing, preventing, or ameliorating a disease, disorder, or side effect, or reducing the rate of progression of a disease or disorder, as compared to a corresponding subject not receiving that amount. The term also includes within its scope dosages effective to enhance normal physiological function. The specific dosage is readily determined by one skilled in the art using routine methods.

The term "combination" as used herein means that therapeutically effective amounts of the constituent drugs are administered simultaneously or sequentially and separately in any manner. Preferably, the drugs are administered within close proximity to each other if not simultaneously. Suitably, both agents are administered within about 24, about 12, about 11, about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, or about 1 hour. As used herein, the administration of bosutinib and T-DM1 is considered to be simultaneous when the dosing interval is less than about 45 minutes.

As used herein, the term "pharmaceutically acceptable carrier and/or excipient" refers to a carrier and/or excipient that is pharmacologically and/or physiologically compatible with the subject and the active ingredient. Pharmaceutically acceptable carriers include, but are not limited to, pH adjusting agents, surfactants, adjuvants, and ionic strength enhancers. For example, pH adjusters include, but are not limited to, phosphate buffered solutions; surfactants include, but are not limited to, cationic, anionic, or nonionic surfactants, such as tween-80; ionic strength enhancers include, but are not limited to, sodium chloride.

As used herein, "subject in need thereof refers to a subject or patient having a disorder or disease associated with overexpression of HER1, HER2, or HER4, or any mutant thereof, which would benefit from administration of a pharmaceutical composition comprising bosutinib and a drug of an anti-HER 1 antibody, an anti-HER 2 antibody, or an anti-HER 4 antibody (e.g., trastuzumab or a drug conjugate thereof such as T-DM 1). These subjects include in particular those patients with HER2 positive breast cancer, metastatic HER2 positive breast cancer or HER2 positive gastric cancer. Suitably, the cancer may be primary breast cancer, that is, a cancer of breast origin. Or the cancer may be secondary, such as metastatic breast cancer, that is, cancer cells metastasize from the breast to secondary sites. Suitably, the method of treating cancer is directed to a subject having gastric cancer, preferably a subject having HER2 positive gastric cancer, more preferably a HER2 positive gastric cancer subject who has been administered one or more chemotherapeutic drugs.

The term "wild-type" as used herein is understood in the art and refers to a polypeptide or nucleotide sequence that occurs in the native population without genetic modification. As is also understood in the art, a "mutant" includes a polypeptide or nucleotide sequence that has been modified at least once with respect to an amino acid or nucleic acid as compared to the corresponding polypeptide or nucleotide sequence present in the wild-type polypeptide or nucleotide sequence, respectively, and the term mutant includes Single Nucleotide Polymorphisms (SNPs) where there is a single base pair difference in the nucleic acid sequence as compared to the most common (wild-type) nucleic acid sequence. Wild-type or mutant forms of HER1, HER2 or HER4 can be identified by known methods; HER1, HER2, or HER4 gene amplification; cancers overexpressing HER1, HER2, or HER4 protein.

The term "antibody" as used herein refers to an immunoglobulin generally consisting of two pairs of polypeptide chains, each pair containing one light (L) and one heavy (H) chain. Antibody light chains can be classified as either kappa or lambda light chains. Heavy chains can be classified as μ, δ, γ, α or ε, and antibody isotypes are defined as IgM, IgD, IgG, IgA and IgE, respectively. In both light and heavy chains, the variable and constant regions are connected by a "J" region of about 12 or more amino acids, and the heavy chain also includes a "D" region of about 3 or more amino acids. Each heavy chain consists of a heavy chain variable region (VH) and a heavy chain constant region (CH). The heavy chain consists of 3 domains (CH1, CH2, and CH 3). Each light chain consists of a light chain variable region (VL) and a light chain constant region (CL). The constant region of the antibody may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component of the classical complement system.

As used herein, the term "antigen-binding fragment" refers to a polypeptide comprising a fragment of a full-length antibody that retains the ability to specifically bind to the same antigen to which the full-length antibody binds, and/or competes with the full-length antibody for specific binding to the antigen.

Pharmaceutical composition

According to the present disclosure, the pharmaceutical composition comprises bosutinib and an anti-HER 1 antibody, an anti-HER 2 antibody or an anti-HER 4 antibody or an antigenic fragment thereof. Suitably, the antibody is selected from trastuzumab, cetuximab, HER4 antibody, MA1-861, HFR1, H4.77.16, an antigenic fragment, or a combination thereof. Suitably, the antibody is selected from trastuzumab, cetuximab or emmetruzumab (T-DM1) and comprises borutinib or any pharmaceutically acceptable salt thereof. Pharmaceutically acceptable salts may include, but are not limited to, salts of inorganic or organic acids. Inorganic acid salts include, for example, salts of hydrochloric acid, hydrobromic acid, sulfuric acid, disulfuric acid, nitric acid, phosphoric acid, perchlorates or bromic acid; organic acid salts may include, for example, salts of formic, acetic, propionic, oxalic, succinic, benzoic, citric, maleic, malonic, malic, tartaric, gluconic, lactic, mandelic, glycolic, pyruvic, glutaric, ascorbic, palmitic, hydroxymaleic, hydroxybenzoic, phenylacetic, cinnamic, methanesulfonic, benzenesulfonic, toluenesulfonic, ethanedisulfonic, gestrinic, fumaric, lactobionic, salicylic, phthalic, pamoic, aspartic, glutamic, rosmarinic, phenylsalicylic, or acetylsalicylic (aspirin).

Administration of a therapeutically effective amount of a pharmaceutical composition of the invention is preferred over administration of a pharmaceutical component alone because the composition provides one or more of the following improvements as compared to administration of a therapeutically effective amount of a pharmaceutical component alone; i) better anti-cancer effect with the single pharmaceutical component having the strongest specific activity, ii) synergistic or highly synergistic anti-cancer activity, iii) provides a dosing regimen that enhances anti-cancer activity and reduces side effects, iv) reduction of toxicity effects, v) increase of therapeutic window, vi) increase of bioavailability of one or more component compounds, or vii) increase of apoptosis compared to the single component compounds.

The invention also relates to the use of a composition of bosutinib and an anti-HER 2 antibody or antigen-binding fragment thereof or a conjugate of bosutinib and said antibody moiety for the manufacture of a kit for detecting the presence or level of HER2 in a sample prior to administering an effective amount of the composition to a patient. The kit optionally includes instructions for detecting HER2 or HER2 levels in a biological sample from the patient.

Method for administering composition

Several malignancies, including lung, breast, gastric, colorectal, head and neck, and pancreatic cancers, are associated with mutations or overexpression of EGFR receptor family members. Bosutinib has therapeutic effects against such cancers, including lung cancer, gastric cancer, breast cancer, and head and neck cancer.

The present disclosure also provides a method of treating cancer comprising administering the pharmaceutical composition to a subject in need thereof. Suitably, a pharmaceutical composition of a therapeutically effective amount of bosutinib and an anti-HER 2 antibody recognizing a HER2 extracellular domain epitope with an antigen affinity constant of up to 0.1nmol/L is administered to a subject in need thereof. Suitably, the anti-HER 2 antibody is a humanized monoclonal antibody, such as trastuzumab, that recognizes a membrane-proximal epitope of HER2 extracellular domain IV with an antigen affinity constant of up to 0.1 nmol/L. Suitably, the antibody recognizes an epitope consisting of 3 loops (557-561, 570-573 and 593-603) at the C-terminus of section IV. Suitably, the antibody may be a humanized bispecific anti-HER 2 antibody or bispecific antigen-binding fragment thereof, said antibody comprising one antigen-binding site comprising the variable regions of the heavy and light chains of trastuzumab, and another antigen-binding site comprising the variable regions of the heavy and light chains. Bispecific antibodies preferably recognize HER2 extracellular domains IV and II. Suitably, the antibody may be a chimeric (mouse/human) monoclonal antibody, for example cetuximab.

Suitably, the pharmaceutical composition is a combination of bosutinib and emrituximab (T-DM 1). The dose of the administration of the bocetitinib is 0.1-50 mg. T-DM1 can be administered in a dose of 0.5-10 mg per kg of patient body weight. Preferably, T-DM1 is administered in an amount of 1.5-5.5 mg per kg body weight. The composition may further comprise oral paclitaxel.

Alternatively, the antibody is cetuximab, an inhibitor of Epidermal Growth Factor Receptor (EGFR), for the treatment of metastatic colon cancer, metastatic non-small cell lung cancer, and head and neck cancer. Cetuximab is a chimeric (mouse/human) monoclonal antibody administered by intravenous infusion under the trade name ErbituxTMSold by the company Michelia, Calif., in the United states and Canada, and merck, Calif., outside of the United states and Canada. In Japan, Merck, Michelia, and Gift company are jointly promoted and sold. As another preparation of the composition of the bosutinib/T-DM 1, cetuximab can be added according to the ratio of 100-500 mg/m2Body surface area administration.

Vinorelbine (NVB) as NavelbineTMIs sold under the trade name of "chemotherapy" and is a drug used in the treatment of a variety of cancers, including breast cancer and non-small cell lung cancer. By intravenous injection or oral administration. Vinorelbine belongs to the alkaloids of the vinca flower. Without being bound by any particular theory, vinorelbine is believed to act by disrupting the normal function of microtubules, thereby preventing cell division. As another preparation of the composition of the bosutinib/T-DM 1, the vinorelbine can be added according to the proportion of 0.5-50 mg/m2Body surface area administration.

Paclitaxel (PTX) in TaxolTMIs sold under the trade name of Kaposi's sarcoma, cancer of the ovary, breast, lung, cervix and pancreas. Administration is by intravenous injection. As another preparation of the bosutinib/T-DM 1 combination, paclitaxel can be added at a ratio of 100-300 mg/m2Body surface area administration.

Suitably, the combination of bosutinib and T-DM1 may further comprise a mitotic inhibitor. The mitotic inhibitor is selected from BT-062, HMN-214, eribulin mesylate, vindesine, EC-1069, EC-1456, EC-531, vindesine (vintafolide), 2-methoxyestradiol, GTx-230, clibulin (crolibulin), D1302A-maytansinoid conjugate, IMGN-529, Moraxel-lovoruzumab (lorvotuzumab mertansine), SAR-3419, SAR-566658, IMP-03138, topotecan/vincristine combination, BPH-8, flubutritin trimethylamine (sbretsulin tromethamine), estramustine sodium phosphate, vincristine, vinflunine, vinorelbine, RX-21101, cabazitaxel, STA-9584, vinblastine A, patupilone (patulopirine), epothilone, paclitaxel, docetaxel, paclitaxel, and the like, DJ-927, discodermolide, eleutherobin and pharmaceutically acceptable salts thereof or combinations thereof. For example, the composition may further comprise a taxane, a vinca alkaloid, or a combination thereof. The vinca alkaloid may be at least one selected from vinblastine, vincristine, vindesine, vinorelbine, and combinations thereof. The taxane may be paclitaxel or docetaxel. Suitably, the combination of bosutinib and T-DM1 may further comprise paclitaxel or vinorelbine. Preferably, the combination of bosutinib and T-DM1 further comprises paclitaxel.

Suitably, the combination of bosutinib and T-DM1 may further comprise an mTOR inhibitor. The mTOR inhibitor is selected from zotarolimus, umirolimus (umirolimus), temsirolimus, sirolimus, and sirolimus nanocrystalTM(NanoCrystalTM) Sirolimus transdermal preparationTM(TransDermTM) sirolimus-PNP, everolimus, biolimusA9, ridaforolimus, rapamycin, TCD-10023, DE-109, MS-R001, MS-R002, MS-R003, Perceiva, XL-765, quinacrine, PKI-587, PF-04691502, GDC-0980, daculisib, CC-223, PWT-33597, P-7170, LY-3023414, INK-128, GDC-0084, DS-7423, DS-3078, CC-115, CBLC-137, AZD-2014, X-480, X-414, EC-0371, VS-5584, PQR-401, PQR-316, PQR-311, PQR-309, PF-06465603, NV-128, nPT-OR, BC-210, WAY-600, WYE-354, WYE-LOR-354, HMPL-518-220, HMYE-518, GNE-317, EC-0565, CC-214, ABTL-0812 and pharmaceutically acceptable salts thereof or compositions thereof. For example, the combination of bosutinib and T-DM1 may further comprise rapamycin. Rapamycin may be in the form of an injection. Rapamycin, also known as sirolimus, is a compound produced by streptomyces hygroscopicus. Rapamil mouldThe content of the extract can be 0.5-10 mg/m2Is administered to the body surface area of (a).

Suitably, the combination of bosutinib and T-DM1 may further comprise an antimetabolite. The antimetabolite may be selected from the group consisting of capecitabine, 5-fluorouracil, gemcitabine, pemetrexed, methotrexate, 6-mercaptopurine, cladribine, cytarabine, doxifluoridine, floxuridine, fludarabine, hydroxyurea, dacarbazine, hydroxyurea, and asparaginase. For example, the composition of bosutinib with T-DM1 may further comprise 5-fluorouracil. The 5-fluorouracil may be in the form of an injection. The 5-fluorouracil can be 100-3000 mg/m2Body surface area administration.

5-Fluorouracil (5-FU) with AdrucilTMIs marketed under the trade name pyrimidine analog, which is used to treat cancer. It can be administered by intravenous injection for treating colon cancer, esophageal cancer, gastric cancer, pancreatic cancer, breast cancer and cervical cancer. As a topical cream for basal cell carcinoma. The mechanism of action is not fully understood, but, without being bound to any particular theory, it is believed to be associated with blocking the action of thymidylate synthase, thereby preventing DNA replication.

Suitably, the combination of bosutinib and T-DM1 may further comprise a platinum antineoplastic agent. The platinum antineoplastic agent is selected from cisplatin, carboplatin, dicycloplatin, eptaplatin, lobaplatin, miboplatin, nedaplatin, oxaliplatin, picoplatin and satraplatin. For example, the bosutinib/T-DM 1 composition may further comprise cisplatin. Cisplatin may be in the form of an injection. Cisplatin can be present in an amount of 1-100 mg/m2Body surface area administration. Without being bound by any particular theory, some of the effects of cisplatin are achieved by binding to DNA, thereby inhibiting DNA replication.

Administering to the human a therapeutically effective amount of a composition of the invention. In general, the therapeutically effective amount of the pharmaceutical formulation of the present invention will depend on a number of factors, such as the age and weight of the subject, the particular condition being treated, the severity of the condition, the nature of the formulation, and the route of administration. Ultimately, the therapeutically effective amount will be determined by the attending physician.

Suitably, the compound is administered simultaneously or sequentially during each treatment cycle. When administered non-concurrently, they are all administered within about 24, about 12, about 11, about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, or about 1 hour, in which case the specified time period is about 24, about 12, about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, about 2, or about 1 hour. As used herein, for a two-component pharmaceutical composition, such as Bozitinib and T-DM1, the dosing interval of Bozitinib and T-DM1 of no more than 45 minutes is considered to be simultaneous dosing.

Suitably, both compounds are administered within a specified time in each treatment cycle of at least 1 day, at least 2 days, at least 3 days, at least 5 days, at least 7 days, at least 14 days, at least 21 days, or at least 30 days, in which case the duration of the treatment cycle is at least 1, 2, 3, 5, 7, 14, 21, or 30 days. If both compounds are administered over a prescribed period of time for more than 30 days during the course of treatment, the treatment is considered long-term treatment and continues until an alteration event occurs, such as a reevaluation of the cancer state or a change in the patient's condition, or one or more serious adverse effects, requiring modification of the regimen.

Suitably, during the course of treatment, for example a pharmaceutical composition of bosutinib and T-DM1, both component drugs are administered for at least 1 day over a specified period of time, followed by administration of bosutinib alone for at least 1, 2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or 29 days, and therefore in this example the treatment period is at least 2-30 days. Suitably, the treatment period is 21 days ± 3 days. Suitably, the treatment period is 21 days.

Also contemplated herein is a drug holiday or drug withdrawal period between the sequential administration of one of the bosutinib and the other drug component and the administration of the other drug. As used herein, a drug holiday (drug withdrawal period) is a period of time after continuous administration of one of the bosutinib and the other drug component, without administration of the bosutinib or the other component, prior to administration of the other drug. Suitably, the drug holiday is selected from the following days: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, and 14 days.

It is understood that each treatment cycle may be followed by one or more repeated dosing cycles, or may be an alternative dosing regimen, with a drug holiday occurring prior to the repeated dosing or alternative dosing regimen.

If the subject has HER2 exon 20 mutant breast cancer, the HER2 exon 20 mutation may comprise a HER2 in-frame exon 20 insertion mutation, a HER2 exon 20 point mutation, or both. The HER2 in-frame exon 20 insertion mutation may be selected from the group consisting of A775_ G776insYVMA, G776_ V777insVC, P780_ Y781insGSP, and combinations thereof. The HER2 exon 20 mutation may be selected from the group consisting of L775S, G776V, V777L, and combinations thereof. The HER2 exon 20 mutation is not a T790M point mutation.

Wild type or mutant HER1, HER2, and HER4 tumor cells can be identified by DNA amplification and sequencing techniques, DNA and RNA detection techniques (including but not limited to Northern and southern blot, respectively), and/or various biochip and array techniques or in situ hybridization. Wild-type and mutant polypeptides can be detected by a variety of techniques, including but not limited to immunodiagnostic techniques such as ELISA, Westernblot, or immunocytochemistry.

Preferably, the method comprises administering to a subject in need thereof a pharmaceutical composition comprising bosutinib and T-DM1, and optionally other chemotherapeutic agents. Preferably, the chemotherapeutic agent comprises paclitaxel.

Suitably, the patient is assessed for HER1, HER2 or HER4 status. It will be appreciated by those skilled in the art that the daily amount of the pharmaceutical composition described herein will be determined at the discretion of the physician within the scope of sound medicine. The specific amount employed in any particular patient will depend upon a variety of factors including the type of cancer being treated and its severity; (ii) the activity of polazitinib in the pharmaceutical composition; the specific pharmaceutical component used; the age, weight, general health, sex, and diet of the subject in need thereof; the administration time is as follows: the prescribed dose for each administration; the duration of the treatment; the drugs in the composition or in combination with the particular pharmaceutical composition used; and similar factors well known in the medical arts.

For example, it is within the skill of the art to start at a dosage that is lower than that required to achieve the desired therapeutic effect and gradually increase the dosage to achieve the desired therapeutic effect. Suitably, the toxicity of a subject receiving the present composition is reduced compared to a subject receiving a standard dose of poecitinib or anti-HER 1 antibody, anti-HER 2 antibody, or anti-HER 4 antibody alone. Suitably, reduced toxicity is associated with reduced cardiotoxicity, such as troponin I rising above 99% above the upper limit of normal range (URL) determined, or any grade 3 cardiovascular toxicity. Suitably, the reduced toxicity is associated with a reduction in hematologic toxicity. For example, subjects receiving combination therapy have a statistically improved rate of platelet to neutrophil depletion compared to subjects receiving drug alone.

Suitably, the subject receiving the combination therapy exhibits an improved and favorable rate of decline in the development of non-rheological toxicity such as diarrhea, rash, mucositis, fatigue, electrolyte or hepatotoxicity.

Another aspect of the invention relates to a method of treating cancer in a subject, comprising administering to the subject a therapeutically effective amount of bosutinib and an anti-HER 1 antibody, an anti-HER 2 antibody, or an anti-HER 4 antibody, wherein the cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER 4. The method may further comprise administering at least one drug selected from paclitaxel, cisplatin, 5-fluorouracil, vinorelbine, cetuximab, or any combination thereof. The antibody is preferably an anti-HER 2 antibody selected from trastuzumab, cetuximab, or any combination of antigen-binding fragments thereof. The cancer may be non-small cell lung cancer, breast cancer, stomach cancer, colon cancer, pancreatic cancer, prostate cancer, myeloma, head cancer, neck cancer, ovarian cancer, esophageal cancer, or metastatic cell cancer.

In at least one embodiment, the method of treating cancer is directed to a subject having breast or gastric cancer.

Suitably, the cancer of interest for the method is breast cancer, wherein the breast cancer is selected from: (i) estrogen receptor negative breast cancer, HER1 and/or HER2 overexpression; (ii) estrogen receptor and progesterone receptor double positive breast cancer, HER2, is expressed but not overexpressed; (iii) trastuzumab-resistant breast cancer, HER2 overexpression; and (iv) triple negative breast cancer, PR, HER2 and estrogen receptor negative, HER1 is overexpressed. The breast cancer is preferably metastatic breast cancer. The method may further comprise collecting breast cancer cells from the subject; breast cancer cells were evaluated to confirm overexpression of HER2, overexpression of HER2 mutant, amplification of HER2, or amplification of HER2 mutant. Suitably, the evaluation step comprises Immunohistochemistry (IHC) and confirmatory Fluorescence In Situ Hybridization (FISH), wherein the IHC may be IHC3+ or IHC2 +.

In one embodiment, the method of treating cancer is directed to a subject with gastric cancer, preferably a subject with HER2 positive gastric cancer, more preferably a HER2 positive gastric cancer subject who has been administered one or more chemotherapeutic drugs.

Another aspect of the invention relates to a method of treating breast cancer in a subject in need thereof, wherein the breast cancer is associated with overexpression or amplification of HER2 or overexpression or amplification of a HER2 mutant, the method comprising a) within a 21 day (± 3 day) cycle, i) single dose administration of T-DM 11.5-5.5 mg/kg; ii) 0.5-50 mg/d of bosutinib is administered daily; and b) optionally repeating said cycle. Suitably, T-DM1 is administered by Intravenous (IV) infusion. Suitably, the standard dose of T-DM1 is 3.6 mg/kg. Suitably, the bosutinib may be administered orally in an oral dose selected from 6, 8,10, 12, 16 and 24mg once daily.

Another aspect of the invention relates to a method of ameliorating the occurrence of an adverse event in a subject undergoing treatment for breast cancer associated with overexpression or amplification of HER2 or a HER2 mutant, the method comprising a) within a 21 day (± 3 day) cycle, i) single dose administration of T-DM 1; ii) administering a daily dose of bosutinib; and b) optionally repeating the cycle, wherein the adverse event is selected from the group consisting of cardiotoxicity, hematologic toxicity, diarrhea, rash, mucositis, fatigue, electrolyte abnormalities, and hepatotoxicity.

Another aspect of the invention relates to a composition for use in the treatment of cancer in a subject, wherein the cancer is associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER4, and the combination comprises therapeutically effective amounts of bosutinib and T-DM1, wherein bosutinib is administered orally and T-DM1 is administered intravenously. The composition may further comprise administering a therapeutically effective amount of paclitaxel by Intravenous (IV) infusion.

Another aspect of the invention relates to a method of treating gastric cancer in a subject in need thereof, wherein gastric cancer is associated with overexpression or amplification of HER2, or overexpression or amplification of a HER2 mutant, and the method comprises the steps of: a) over a 21 day (± 3 days) cycle; i) the trastuzumab is administered in a single dose of 6-8 mg/kg; ii) a single dose of paclitaxel at 105-175 mg/m2(ii) a iii) administering 4-16 mg/d of bosutinib per day; b) the above cycle is optionally repeated. Trastuzumab is preferably administered by Intravenous (IV) infusion on day 1. Preferably, the standard dose of trastuzumab is an 8mg/kg loading followed by an infusion of 6 mg/kg. Paclitaxel is preferably administered by Intravenous (IV) infusion on day 1. The standard dose of paclitaxel is preferably 175mg/m2. The bosutinib is orally administered for 14 + -3 days at a dose selected from 4, 6, 8,10, 12 and 16mg once daily. A suitable drug holiday 14 days after administration of bosutinib was 7 days.

The technical solutions provided by the present invention are described in detail below with reference to examples, but they should not be construed as limiting the scope of the present invention.

Example 1: methods of treating HER2 positive breast cancer

Generally, a female patient having breast cancer is identified, baseline imaging is performed, breast tissue is obtained for tumor genotyping, the patient undergoes a treatment period based on tumor typing, the treatment regimen is evaluated and optionally continued or adjusted.

The presence of HER2 overexpressing or gene amplified tumors in patients was confirmed by Fluorescence In Situ Hybridization (FISH) by Immunohistochemistry (IHC) labeling of IHC3+ or IHC2 +. The IHC test provides a score of 0 to 3+ for measuring the amount of cell surface HER2 receptor protein in breast cancer tissue samples. If the score is 0 to 1+, then the tissue sample is considered "HER 2 negative"; if the score is 2+, then the tissue sample is considered "critical"; tissue samples scored 3+ were considered "HER 2 positive"

The patients are treated according to a cycle of 21 days +/-3 days, 3.6mg/kg of T-DM1 is intravenously dripped on day 1, and the bosutinib is orally taken on days 1-21, wherein the dosage is 6mg, 8mg, 10mg, 12mg or 16mg every day. The high dose of bosutinib (24 mg per day) may be administered orally, but the dosing regimen includes a 1-week drug holiday. 24mg/d is given on days 1-14, and then taken out once on days 15-21 of a 21-day cycle. The cycle was repeated for 21 days until disease progression, death or intolerable Adverse Events (AEs) occurred.

A phase 1b open label multicenter study was performed on patients with advanced or metastatic HER2 positive breast cancer to determine the Maximum Tolerated Dose (MTD) or the Maximum Administered Dose (MAD) of boresinib at the time of standard dose of T-DM 1. The dose of bocetinib determined in part 1 of the study was used in combination with the standard starting dose of T-DM1(3.6mg/kg i.v. on day 1 of each 21 day cycle) in part 2 for efficacy assessment.

Thus, the first part of the study is dosimetry. The first group was orally administered 8mg of bosutinib initially and on day 1 of each cycle in combination with a standard starting dose of T-DM1(3.6mg/kg i.v.), followed by 8mg of bosutinib per day. The dose escalation of azitinib was performed according to the "3 + 3" design, based on the occurrence of dose-limiting toxicity (DLT) in cycle I of the current dose group.

DLT refers to any treatment-related toxicity that occurs during the first treatment cycle:

cardiotoxicity:

troponin I was elevated to 99% above the normal range tested or above the Upper Reference Limit (URL) for any grade 3 cardiovascular toxicity

Blood toxicity:

grade 3 thrombocytopenia with bleeding;

grade 4 neutropenia >7 days or grade 3 febrile neutropenia

Non-hematologic toxicity:

grade 3 or above grade 3 diarrhea for >3 days, or with fever above 100.5 ° F (38.1 ℃) for at least 2 days or severe dehydration;

grade 4 rash or mucositis;

grade 3 or above grade 3 fatigue for at least 1 week;

according to hepatotoxicity evidence of Hai's law (ALT or AST is more than or equal to 3 × ULN, total bilirubin is more than 2 × ULN, no viral hepatitis, liver disease or liver metastasis; patients with liver metastasis, the duration is more than or equal to 14 days ALT or AST is more than 8 × ULN or AST or ALT is more than 5 × ULN;

grade 3 or more electrolyte abnormalities with a duration of more than 72 hours, unless the patient has clinical symptoms, in which case all grade 3 or more electrolyte abnormalities, regardless of duration, should be counted as DLT; an increase in grade 3 or more amylase or lipase, independent of pancreatitis symptoms or clinical manifestations, need not be counted as DLT;

any other non-hematologic toxicity associated with treatment no less than grade 3;

death from an undefined underlying disease or external cause.

Toxicity assessments were based on the grade of adverse events, performed using the National Cancer Institute (NCI) international common adverse reaction standard (CTCAE), version 4.03.

In phase 1b part 1, the initial dose of bosutinib in combination with T-DM1(3.6mg/kg i.v.) was determined using a "3 + 3" design at each cycle, with a maximum of 3 dose levels tested starting at 8 mg/day. The dose escalation of the azitinib was performed for the next dose group based on the occurrence of dose-limiting toxicity (DLT) in cycle I of the current dose group. Patients who completed cycle I continued treatment at this set of doses until treatment was discontinued. Four possible dose groups included 8,10 and 12mg/d of bozitinib, plus 6mg/d of bozitinib reduced dose groups.

In part 2 of the phase 1b study, approximately 10 patients were treated with MTD/MAD to determine the safe dose of the combination and to assess the primary efficacy. Treatment of all patients continues until the disease has progressed, unacceptable toxicity or continued treatment is considered not beneficial to the patient.

In each 21-day cycle, eligible patients received a prescribed dose of bosutinib orally, once a day, and were given a glass of water and breakfast continuously in the morning at approximately the same time. On day 1 of each treatment cycle, 3.6mg/kg of T-DM1 was injected intravenously. On the day of T-DM1 administration, bosutinib was administered sequentially after the end of the infusion.

After 3 treatment cycles, tumor remission was assessed every 9 weeks ± 14 days.

Example 2: methods of treatment, dosage regimens, efficacy and safety/tolerability of HER2 positive breast cancer

Dose regimens were formulated according to cycle II, open label multicenter studies and evaluated for initial efficacy and safety/tolerability of bosutinib against HER2 positive Metastatic Breast Cancer (MBC) patients who received at least a targeted treatment regimen comprising both trastuzumab and T-DM1 HER 2. Bozitinib is administered at a dose of 24mg/d (1 week every 2 weeks of drug holidays), 16mg/d (continuous daily dosing) or 12mg/d (continuous daily dosing) over a period of 21 days. Toxicity assessments were based on the grade of adverse events, performed using the National Cancer Institute (NCI) international common adverse reaction standard (CTCAE), version 4.03.

With respect to combination therapy further including another drug, a high degree of synergy was observed when bosutinib was used in combination with paclitaxel, cisplatin or 5-FU for breast cancer cells BT-474 cells that overexpress HER2 and are Estrogen Receptor (ER) negative. When bosutinib is used in combination with trastuzumab, GI at certain concentrations equal to or lower than that of bosutinib alone50When it is used, a weak synergistic effect is exhibited, wherein GI50Is the drug concentration that results in a 50% reduction in cancer cell proliferation. GI at a concentration equal to or lower than that of paucitinib alone when used in combination with vinorelbine50No synergy was observed. Similarly, a synergistic effect was observed when vinorelbine was applied in combination with poetinib to HER2 overexpressing and ER negative SK-BR-3 cells. However, the combination of bosutinib and vinorelbine showed synergistic effect at certain concentrations when applied to HER2 over-expressed, ER positive and trastuzumab resistant MDA-MB-361 cells; and the combined application of the bosutinib and the vinorelbine to MCF-At 7 cells, each concentration showed a synergistic effect, whereas MCF-7 cells were ER positive and HER1 and HER2 were not overexpressed. In addition, a synergistic effect was also observed at certain concentrations when bosutinib and vinorelbine were applied in combination to HER2 negative, ER negative and HER 1-overexpressed triple negative breast cancer cells MDA-MB-468 cells. Paclitaxel, 5-FU, cisplatin or trastuzumab in combination with Boletinib for MBA-MB-453 cells of breast cancer cells over-expressed HER2 and resistant to trastuzumab, at concentrations equal to or lower than GI of Boletinib alone50A significant synergistic effect was observed.

When the bosutinib is combined with 5-FU in TE cells (an esophageal cancer cell line with over-expression of HER 2), the synergistic effect of the combination of the bosutinib and other medicines is very good. In addition, when bosutinib was used in combination with trastuzumab in the gastric cancer cell line N-87 cells with overexpression of HER2, synergy was observed at certain concentrations.

Therefore, the composition of bosutinib and T-DM1, further comprising other targeted anticancer agents or cytotoxic anticancer agents, preferably treats cancers associated with overexpression or amplification of HER1, HER2, HER4, a mutant of HER1, a mutant of HER2, or a mutant of HER4, is very effective in cancers such as breast cancer, gastric cancer, lung cancer, and esophageal cancer, and is effective in inhibiting cancers resistant to conventional therapeutic drugs. Suitably, the cancer is HER2 positive breast cancer.

Example 3: treatment of advanced gastric cancer with HER2 positive

A prospective phase I/II study was conducted at 11 treatment centers in korea. HER2 positive gastric cancer patients who received one or more chemotherapeutic agents were enrolled. The patients took bosutinib (8mg or 12mg) orally daily for 14 days, followed by drug holidays for 7 days. Paclitaxel (175 mg/m)2Infusion) and trastuzumab (8mg/kg loading dose followed by 6mg/kg infusion) were co-administered with bosutinib on day 1 every 3 weeks.

Method of producing a composite material

The patients: the recruitment criteria mainly include: patients aged 19 years or more; histopathology confirmed local eveningUnresectable, recurrent, or metastatic gastric adenocarcinoma (including esophageal-gastric junction adenocarcinoma); HER2 Immunohistochemistry (INC)3+ or HER2 IHC2+ and HER2 Fluorescence In Situ Hybridization (FISH) +; assessing whether one or more measurable lesions are present in the solid tumor according to a therapeutic efficacy assessment criteria (RECIST, version 1.1) for the solid tumor; regardless of the exposure history of trastuzumab, chemotherapy drugs used include fluoropyrimidine or platinum; and adequate bone marrow and liver function. Key exclusion criteria included: is provided withA history of drug allergy to EL and trastuzumab; previous exposure to taxanes, and symptomatic central nervous system metastases.

The characteristics of the patients are as follows: a total of 44 patients (12 stage I, 32 stage II) were from 11 different treatment centers in korea. Table 1 summarizes the basic characteristics of the study population.

TABLE 1 summary of basic characteristics of study population

The study was conducted in accordance with the declaration of helsinki and international conference on coordination (ICH) on clinical trial administration protocol (GCP) and was approved by the institutional review board of each participating treatment center. All patients provided written informed consent prior to enrollment.

The treatment procedure is as follows: each 21-day cycle included oral administration of bosutinib once daily for 14 days, and paclitaxel (175 mg/m) on day 12Infusion) and trastuzumab (8mg/kg loading dose followed by 6g/kg infusion). In the study of item IIn phase, the dose of bosutinib (8mg, 12mg or 16mg) was increased, presuming RP2D for the combination of bosutinib with paclitaxel and trastuzumab. At each dose level, DLT was assessed in 6 patients. If DLT is observed in ≦ 1 patient, the dose continues to increase to the next dose level. If DLT is observed in 2 patients ≧ 2, the dose escalation is discontinued and the MTD is determined as the highest level of DLT occurring in 1 patient ≦ 1. In cycle II of the study, 32 patients were treated with RP2D pazetinib in combination with paclitaxel and trastuzumab.

Safety analysis and efficacy: adverse Events (AEs) and adverse events occurring during Treatment (TEAEs) were evaluated using the national cancer institute adverse event general terminology standard (version 4.03). DLT is defined as grade 3 non-hematologic toxicity (except alopecia); despite the maximum dose of anti-diarrheal and/or antiemetic agent, diarrhea, nausea and vomiting are grade 3 or greater (as applicable); grade 4 neutropenia lasts for more than or equal to 7 days, and grade 3-4 neutropenia is accompanied by fever or infection; grade 4 thrombocytopenia and grade 3 thrombocytopenia, lasting 7 days, with bleeding or the need for transfusion. Left Ventricular Ejection Fraction (LVEF) basal assessment was performed on day 1 of the first 3 cycles and every 3 cycles thereafter. All patients receiving at least one treatment with breccitinib were included in the safety analysis. CT or MRI evaluations were performed on the chest, abdomen and pelvis every 6 weeks using RECIST version 1.1, according to investigator's evaluations.

Statistical analysis: the primary objective of cycle I was to assess the safety and tolerability of pozzotinib and to determine the MTD of pozzotinib when administered in combination with paclitaxel and trastuzumab. The primary objective of cycle II was to assess the Objective Remission Rate (ORR) of bociclib in combination with paclitaxel and trastuzumab. Secondary objectives were to evaluate safety and tolerability, Progression Free Survival (PFS), time to disease progression (TTP) and overall response Duration (DOR).

For cycle II, ORR > 20% was considered clinically significant assuming ORR < 5% ineffective. The sample size was calculated using Simon two-stage maxima and minima, 80% effective rate and 5% significance level. Of the 27 subjects targeted, 13 subjects were required for cycle I. If tumor remission was 0 out of 13 patients, the study design was terminated prematurely. However, if at least 1 response was observed, then an additional 14 patients will be included in the study.

Results

Period I

7 grade 1 (8mg of poecitinib) dose patients were enrolled, but 1 of them failed to assess DLT due to incomplete follow-up before the end of the I-th cycle. Of the remaining 6 patients, 1 (grade 4 neutropenia) developed DLT. At a grade 2 dose level (12 mg of bosutinib), 5 patients were enrolled, and 2 patients observed to develop DLT (grade 4 neutropenia in one patient and febrile granulocytopenia with grade 4 neutropenia in another patient). Therefore, according to the above criteria, the bovatinib MTD was determined to be 8 mg.

Toxicity and dose adjustment. Table 2 summarizes the safety analysis data set for 12 patients with phase I TEAE. All 12 patients (100%) experienced ≧ 1 TEAE, of which 11 patients (91.7%) were observed for grade 3 toxicity. 3 patients (25.0%) presented TEAE associated with study discontinuation. There were 1 case (8.3%) of TEAE that caused death (see table 2). The most common boresinib-related adverse events were diarrhea, rash, stomatitis, itching and anorexia (table 3). Grade 3 or higher neutropenia and febrile neutropenia were observed in 9 (75%) and 2 (16.7%) patients, respectively (supplementary table 1). 1 of 6 patients (14.3%) received a dose reduction (from 8mg to 6mg) of bosutinib. Paclitaxel dose reduction and drug withdrawal were performed in 2 (28.6%) and 1 (14.3%) patients, respectively (supplementary table 2).

TABLE 2 comprehensive summary of Treatment Emergent Adverse Events (TEAE)

Table 3 security: adverse events associated with Pocetinic acid (. gtoreq.10% of patients)

Supplementary Table 1 incidence of most common (≧ 10%) and ≧ 3 class of TEAE for bosutinib

Supplementary table 2 dose adjustments

The efficacy is as follows: a total of 11 patients were available to evaluate the efficacy of boresinib. Objective remission was observed in 4 patients (33.3%; 95% CI, 9.9-65.1) (Table 4). There was partial relief in all four. The disease control rate (DCR; PR + SD) was 66.7% (95% CI, 34.9-90.1). Median survival and median overall survival were 17.7 weeks (95% CI, 5.4-30.2) and 30.6 weeks (95% CI, 12.2-195.0), respectively (table 5 and fig. 1).

TABLE 4 summary of tumor remission

TABLE 5 summary of survival data

CI is the confidence interval. If the last dosing date was not collected, the study end date is used. The number of patients receiving 8mg and 12mg treatment in cycle I was 2 and 2, respectively. Cycle II (8mg) 7 patients. Based on the duration of the overall reaction, a total of 11 cases were calculated.

Period II-see table above.

A total of 32 patients entered cycle II. 20 patients (63%) received low doses of bosutinib. See supplementary table 2 above.

And (4) safety analysis: TEAE neutropenia and febrile neutropenia of grade 3 or higher were seen in 4 (12.5%) and 3 (9.4%) patients, respectively (supplementary table 1). The most common poezinib-associated TEAEs were diarrhea, rash, stomatitis and decreased appetite, similar to the results of cycle I (table 3). 13 patients (40.6%) developed grade 3 or higher diarrhea (supplementary table 1). 4 patients (12.5%) stopped the study before first evaluation due to treatment-related toxicity. No grade 3,4 Left Ventricular Systolic Dysfunction (LVSD). 20 (62.5%) patients received low dose treatment with bosutinib. Paclitaxel was given at reduced doses in 12 patients (37.5%) and was discontinued in 3 patients (9.4%) (supplementary table 2).

The efficacy is as follows: in 20 (62.5%) patients, tumor size was reduced (FIG. 1). Confirmed responses were observed in 7 patients (21.9%; 95% CI, 9.3-40.0). 2 cases (6.3%) were in complete remission and 5 cases (15.6%) were in partial remission. The disease control rate was 71.9% (95% CI, 53.3-86.3) (Table 4). The average PFS was 13.0 weeks (95% C, 9.8-21.9) (Table 5, FIGS. 1 and 3). The median overall survival was 29.5 weeks (95% CI, 17.9-59.2) (table 5, fig. 1). The TTP and DOR medians were 15.0 weeks (95% CI, 10.0-23.1) and 26.8 weeks (95% CI, 17.2-71.8), respectively (Table 5). Objective remission rates were 12.5% and 50.0% for trastuzumab pretreated and non-trastuzumab treated patients, respectively, when the clinical efficacy was studied based on trastuzumab exposure (table 4). The median overall survival and median survival for trastuzumab-pretreated patients were 29.5 weeks (95% CI, 17.9-40.9) and 13.0 weeks (9.8-18.7), respectively, and for trastuzumab-treated patients were 42.6 weeks (95% CI, 3.0-111.5) and 18.7 weeks (95% CI, 3.0-) (table 5).

Discussion of the related Art

In this prospective, open label multicenter, cycle II study, 8mg of bociclib in combination with paclitaxel and trastuzumab treated patients with advanced HER2 positive Gastric Cancer (GC) showed controlled toxicity and good efficacy. To our knowledge, this study evaluated for the first time the efficacy of pan-HER inhibitors in combination with chemotherapeutic drugs and trastuzumab for the treatment of HER2 positive tumors.

In the previous cycle I study, the MTD of Boletinib was determined to be 24mg/d (14 days with intermittent dosing and 7 days with rest) and 18mg/d (with continuous dosing). In cycle I of the study, the MTD of poecitinib was determined to be 8mg/d, which is much lower than the 24mg/d MTD of poecitinib administered alone. Common TEAEs are diarrhea, rash, stomatitis and itching. In this study, all patients developed at least one TEAE, with a similar incidence as reported in the cycle I trial of the monotherapy with bosutinib. Unlike the cycle I monotherapy trial, all DLTs in our study were grade 4 neutropenia or febrile neutropenia with grade 4 neutropenia, rather than diarrhea.

In the above-described cycle I trial, objective remission and median survival for treatment with bozitinib monotherapy were 16% and 12.0 weeks (intermittent dosing regimen) and 21% and 9.0 weeks (continuous dosing regimen), respectively. Another pan-HER inhibitor, dacomitinib (dacomitinib), produced an objective remission rate of 7.4% (95% CI, 0-17.5) and a median survival of 2.1 months (95% CI, 2.3-3.4) in HER2 positive gastric cancer as monotherapy. Based on the objective remission rates and median survival of our studies, these results suggest that pan-HER inhibitor combination chemotherapy may be more effective than pan-HER inhibitor alone.

Double blockade of the HER2 receptor has become the standard therapy for HER2 positive breast cancer. Pertuzumab in combination with docetaxel and trastuzumab significantly increased the overall survival of HER2 positive breast cancer. However, pertuzumab combined chemotherapy did not prolong the overall survival of HER2 positive gastric cancer compared to HER2 positive breast cancer. This finding suggests that the efficacy of the dual blocker on HER2 positive gastric cancer may be different from HER2 positive breast cancer. However, even in vitro, the dual blockers of lapatinib and trastuzumab show highly synergistic anti-tumor activity against HER 2-expanded gastric cancer cells. In the current study, bosutinib in combination with trastuzumab showed good tumor remission, suggesting that this dual blocking strategy, including pan-HER inhibitors, would be a promising therapeutic option for HER2 positive gastric cancer rescue treatment.

In HER2 positive breast cancer, continued anti-HER 2 targeted therapy is a recognized therapeutic strategy. A retrospective study report reported that trastuzumab increased progression-free survival and overall survival of HER2 positive gastric cancer. However, in a prospective cycle II study, the failure of non-progressive trastuzumab as a second-line drug to improve progression-free survival and objective remission rates in HER 2-positive gastric cancer patients suggests that the role of non-progressive trastuzumab in HER 2-positive gastric cancer patients remains controversial.

Antibody-drug conjugates (ADCs) are one of the new methods for treating HER2 positive tumors. The enzmetuzumab cycle 3 trial was performed in HER2 positive gastric cancer patients who previously failed treatment with fluoropyrimidine and platinum-based drugs. However, emmetruzumab is not superior to paclitaxel. The objective remission rate was only 20.6% (95% CI, 15.26-26.45). DS8201a is a novel ADC targeting HER2 and has been shown to have strong antitumor activity against HER2 positive gastric cancer cells. In the cycle I study, the objective remission rate of DS8201a for trastuzumab-resistant HER2 positive gastric cancer patients and low HER2 expressing gastric cancer patients was 44% and the disease control rate was 78%, indicating that HER 2-targeted ADC is a promising rescue treatment in HER2 positive gastric cancer.

In this study, pre-conditioning biopsies were not mandatory, so patients were recruited based on HER2 expression at the time of initial diagnosis. However, HER2 positive gastric cancer patients differed in HER2 expression before and after trastuzumab treatment, with 32% of patients receiving trastuzumab even observing positive inactivation of HER 2. These observations suggest that it is necessary to re-assess the exact HER2 status before again targeting therapy with anti-HER 2 and therefore this is a limitation of our study.

In summary, the combined use of bosutinib (8mg) with paclitaxel and trastuzumab has good clinical efficacy and controllable toxicity for HER2 positive gastric cancer patients receiving first-line chemotherapy. Dual blockade inhibition of trastuzumab with pan-HER inhibitors is a promising strategy to overcome trastuzumab resistance.

It should be understood that the embodiments described herein are to be considered merely illustrative and not restrictive of the scope of the invention, and that the description of features or aspects of each embodiment should generally be regarded as other similar features or aspects that may be used in other embodiments. It will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the disclosure as defined by the appended claims.

30页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:新型抗分枝杆菌杂环酰胺

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!