CN116583540A - Combination therapy for the treatment of HER2 cancer - Google Patents

Combination therapy for the treatment of HER2 cancer Download PDF

Info

Publication number
CN116583540A
CN116583540A CN202180083440.6A CN202180083440A CN116583540A CN 116583540 A CN116583540 A CN 116583540A CN 202180083440 A CN202180083440 A CN 202180083440A CN 116583540 A CN116583540 A CN 116583540A
Authority
CN
China
Prior art keywords
administered
day
trastuzumab
pertuzumab
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180083440.6A
Other languages
Chinese (zh)
Inventor
J·L·舒茨曼
C·宋
K·W·宋
E·塔诺普卢
S·P·华伯顿
A·戴伊
S·R·朱
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Genentech Inc
Original Assignee
F Hoffmann La Roche AG
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F Hoffmann La Roche AG, Genentech Inc filed Critical F Hoffmann La Roche AG
Priority claimed from PCT/US2021/062101 external-priority patent/WO2022125483A1/en
Publication of CN116583540A publication Critical patent/CN116583540A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application provides combination therapies for treating HER2 positive cancers comprising inanolisib (also known as GDC-0077) and other HER2 targeted therapies (e.g., pertuzumab and trastuzumab); and methods of treating HER2 positive (her2+) cancer in a patient, preferably a patient having PIK3CA mutant breast cancer, comprising administering therapeutically effective amounts of inanolisib and HER2 targeted therapies (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab).

Description

Combination therapy for the treatment of HER2 cancer
Cross Reference to Related Applications
The present application claims priority from U.S. provisional patent application number 63/124,495 filed on 11 of 12 months in 2020, U.S. provisional patent application number 63/161,153 filed on 15 of 3 months in 2021, and U.S. provisional patent application number 63/209,302 filed on 10 of 6 months in 2021; the contents of these U.S. provisional applications are incorporated by reference herein in their entirety.
Technical Field
The present application relates generally to the treatment of locally advanced or metastatic PIK3CA mutated solid tumors, including HER2 positive breast cancer, in a patient by administering the PI3K inhibitor inanolisib (also known as GDC-0077) in combination with other HER2 targeted therapies, such as pertuzumab (pertuzumab) and trastuzumab (trastuzumab).
Background
Worldwide, breast cancer is the second most common invasive malignancy and the most common cause of cancer-related death in females, with a 5-year survival rate of about 15% after metastatic diagnosis.
Phosphatidylinositol 3 kinase (PI 3K) is a lipidPlasma kinases, upon activation by growth factor receptors and integrins, can regulate proliferation, survival, and migration of cells. PI3K catalyzes phosphatidylinositol 4, 5-bisphosphate (PIP) 2 ) To produce phosphatidylinositol-3, 4, 5-triphosphate (PIP) 3 ) This is a second messenger involved in phosphorylation of AKT and other components of the AKT/mTOR pathway. Up to 70% of breast cancers have some form of molecular aberration of the PI3K/AKT/mTOR pathway. Activating mutations in PIK3CA encoding the PI3Kp110 alpha subunit are very common in breast cancer and solid tumor malignancies.
While PI3K alpha inhibitors have been approved or in the clinical development stage for the treatment of Hormone Receptor (HR) positive, HER2 negative, locally advanced or metastatic breast cancer patients with PIK3CA mutations, there remains a need for agents for the treatment of HER2 positive cancers.
Disclosure of Invention
The present disclosure provides a combination therapy comprising inanolisib (GDC-0077) and HER2 targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab) for treating HER 2-overexpressed breast cancer.
One aspect of the present disclosure provides a combination therapy comprising inanolisib, trastuzumab, and pertuzumab for treating a PIK3CA mutated HER2 positive (her2+) breast cancer.
The present disclosure further provides a method for treating locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer, the method comprising administering to a patient in need thereof a therapeutically effective amount of inanolisib or a pharmaceutically acceptable salt thereof, and HER2 targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab).
In one aspect, the present disclosure provides a method for treating HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to a patient in need thereof a therapeutically effective amount of inanolisib or a pharmaceutically acceptable salt thereof, trastuzumab and pertuzumab.
In one aspect, the present disclosure provides a method for treating HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to a patient in need thereof a therapeutically effective amount of inanolisib or a pharmaceutically acceptable salt thereof, trastuzumab, pertuzumab, and paclitaxel.
In some embodiments, the patient has HER2+ breast cancer with locally advanced or metastatic PIK3CA mutations. In some embodiments, the patient has a Left Ventricular Ejection Fraction (LVEF) of 50% or higher. In some embodiments, the patient is female.
In some embodiments, the patient has HER2 positive breast cancer with a hormone receptor positive (hr+) locally advanced or metastatic PIK3CA mutation, and the method further comprises administering an endocrine therapy (e.g., fulvestrant or letrozole) to the patient.
Also provided is a combination for treating locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer, wherein the combination comprises inanolisib or a pharmaceutically acceptable salt thereof and HER2 targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab).
Also provided is the use of a combination in the manufacture of a medicament for the treatment of locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer, wherein the combination comprises inanolisib or a pharmaceutically acceptable salt thereof and HER2 targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab).
In some embodiments, inavelisib (GDC-0077) is administered at a daily dose of 3mg, 6mg, or 9 mg. In one embodiment, inanolisib (GDC-0077) is administered orally at a daily dose of 9 mg.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising: inavilisib, trastuzumab and pertuzumab; wherein the combination therapy is administered over a 21 day period.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. QDs were administered inavilisib on days 1 to 21 of the first 21 day cycle;
b. trastuzumab was administered on day 1 of the first 21-day cycle; and
c. pertuzumab was administered on day 1 of the first 21 day cycle.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 2 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle;
d. pertuzumab was administered on day 2 of the first 21 day cycle.
In some embodiments, the dosing regimen further comprises up to five additional 21-day periods, the five additional 21-day periods comprising:
e. Paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
f. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
g. trastuzumab was administered on day 1 of each additional 21-day cycle; and
h. pertuzumab was administered on day 1 of each additional 21-day cycle.
In some of these embodiments, the method further comprises one or more additional 21-day cycles comprising:
a. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
b. trastuzumab was administered on day 1 of each additional 21-day cycle; and
c. pertuzumab was administered on day 1 of each additional 21-day cycle.
Also provided is a combination for treating HER2 positive breast cancer in a patient suffering from locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer, wherein the combination comprises inavelisib, trastuzumab, pertuzumab optionally paclitaxel; wherein the combination therapy is administered over a 21 day period.
Also provided is a combination for treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, wherein the combination is administered as a combination therapy comprising a dosing regimen comprising:
a. QDs were administered inavilisib on days 1 to 21 of the first 21 day cycle;
b. trastuzumab was administered on day 1 of the first 21-day cycle; and
c. pertuzumab was administered on day 1 of the first 21 day cycle.
Also provided is a combination for treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, wherein the combination is administered as a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 2 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle;
d. pertuzumab was administered on day 2 of the first 21 day cycle.
In some embodiments, the dosing regimen further comprises up to five additional 21-day periods, the five additional 21-day periods comprising:
e. paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
f. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
g. trastuzumab was administered on day 1 of each additional 21-day cycle; and
h. pertuzumab was administered on day 1 of each additional 21-day cycle.
Also provided is the use of a combination in the manufacture of a medicament for treating HER2 positive breast cancer in a patient suffering from locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer, wherein the combination comprises inavillisib, trastuzumab, pertuzumab and optionally paclitaxel; wherein the combination therapy is administered over a 21 day period.
Also provided is a use of a combination in the manufacture of a medicament for treating HER2 positive breast cancer in a patient suffering from HER2 positive breast cancer having a locally advanced or metastatic PIK3CA mutation, wherein the combination is administered in a combination therapy comprising a dosing regimen comprising:
a. QDs were administered inavilisib on days 1 to 21 of the first 21 day cycle;
b. trastuzumab was administered on day 1 of the first 21-day cycle; and
c. pertuzumab was administered on day 1 of the first 21 day cycle.
Also provided is a use of a combination in the manufacture of a medicament for treating HER2 positive breast cancer in a patient suffering from HER2 positive breast cancer having a locally advanced or metastatic PIK3CA mutation, wherein the combination is administered in a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 2 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle;
d. pertuzumab was administered on day 2 of the first 21 day cycle.
In some embodiments, the dosing regimen further comprises up to five additional 21-day periods, the five additional 21-day periods comprising:
e. paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
f. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
g. trastuzumab was administered on day 1 of each additional 21-day cycle; and
h. pertuzumab was administered on day 1 of each additional 21-day cycle.
In some of these embodiments, the dosing regimen further comprises one or more additional 21-day periods comprising:
a. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
b. trastuzumab was administered on day 1 of each additional 21-day cycle; and
c. pertuzumab was administered on day 1 of each additional 21-day cycle.
In some of these embodiments, inanolisib is administered in an amount of 9mg, for example, in an oral tablet. In some embodiments, trastuzumab is administered by Intravenous (IV) infusion at a load dose of 8mg/kg during the first 21-day period and at a dose of 6mg/kg during each additional 21-day period. In some embodiments, pertuzumab is administered by IV infusion at a loading dose of 840mg during the first 21 day period and at a dose of 420mg during each additional 21 day period. In some embodiments, paclitaxel is further infused by IV at 80mg/m over the first 21 day period and up to five 21 day periods 2 Is administered in a weekly dose.
In some embodiments, the patient has HER2 positive breast cancer with hormone receptor positive (hr+) locally advanced or metastatic PIK3CA mutations. In some of these embodiments, the method further comprises administering fulvestrant (e.g., about once every four weeks at a dose of 500mg by intramuscular infusion) or letrozole (e.g., once daily at a dose of 2.5mg in an oral tablet) to the patient.
In yet another aspect, a method of inhibiting tumor growth or producing/increasing tumor regression in a patient suffering from locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer is provided, the method comprising administering a combination therapy to the patient according to the methods detailed herein.
In another aspect, there is provided a combination for inhibiting tumor growth or producing/increasing tumor regression in a patient suffering from HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutation, for use in accordance with the combination detailed herein.
In another aspect, there is provided a use of a combination in the manufacture of a medicament for inhibiting tumor growth or producing/increasing tumor regression in a patient suffering from HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutation, according to the combination or use detailed herein.
Further provided is a method of preventing or delaying the development of resistance of a tumor (e.g., breast cancer) to a therapy comprising HER 2-targeted therapy, the method comprising administering a combination therapy comprising inanolisib, trastuzumab, and pertuzumab. In some embodiments, the combination therapy is administered according to any of the methods as detailed herein.
Also provided is a combination for preventing or delaying the development of resistance of a tumor (e.g., breast cancer) to a therapy comprising HER 2-targeted therapy, wherein the combination comprises inavelisib, trastuzumab, pertuzumab, and optionally paclitaxel. In some embodiments, the combination is administered according to any use as detailed herein.
Also provided is the use of a combination in the manufacture of a medicament for preventing or delaying the development of resistance of a tumor (e.g., breast cancer) to a therapy comprising HER2 targeted therapy, wherein the combination comprises inanolisib, trastuzumab, pertuzumab, and optionally paclitaxel. In some embodiments, the combination is administered according to any use as detailed herein.
Drawings
FIG. 1 shows viability IC determined by quantifying ATP from breast tumor cell lines 50 Value: HER2 positive PIK3CA mutant (n=6), HER2 positive PIK3CA wild type (n=4), HER2 negative PIK3CA mutant (n=10) and HER2 negative PIK3CA wild type (n=20) for 5 days, the concentration of inaviolisib or apersib was increased.
Fig. 2 shows tumor growth curves from KPL-4 (her2+, PIK3CA H1047R) xenografts, which were treated with either the xenograft vehicle, inanolisib, trastuzumab+pertuzumab, or combination therapy of inanolisib, trastuzumab and pertuzumab.
Detailed Description
Definition of the definition
The words "comprise (comprise, comprising)" and "comprising (include, including)" when used in this specification and the claims are intended to specify the presence of stated features, integers, components, or steps, but they do not preclude the presence or addition of one or more other features, integers, components, steps, or groups thereof.
The term "treatment" refers to therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as the growth, development or spread of cancer. For the purposes of the present invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "treatment" may also mean an increase in survival compared to the expected survival without treatment. The person in need of treatment includes those already with the condition or disorder, as well as those prone to have the condition or disorder or those for whom the condition or disorder is to be prevented.
The phrase "therapeutically effective amount" refers to an amount of a compound of the invention that (i) treats a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder as described herein. In the case of cancer, a therapeutically effective amount of the drug may reduce the number of cancer cells; reducing tumor size; inhibit (i.e., slow or preferably stop to some extent) infiltration of cancer cells into peripheral organs; inhibit (i.e., slow or preferably stop to some extent) tumor metastasis; inhibit tumor growth to some extent; and/or to some extent, alleviate one or more of the symptoms associated with the cancer. To some extent, the agent may prevent growth and/or kill existing cancer cells, which may be cytostatic and/or cytotoxic. For cancer therapy, efficacy may be measured, for example, by assessing time to disease progression (TTP) and/or determining Remission Rate (RR).
"time of progression" or "TTP" refers to the time since randomization to target tumor progression.
"objective remission rate" or "ORR" refers to the proportion of patients identified by researchers as fully or partially remitted during two consecutive periods of greater than or equal to 4 weeks apart, as determined by RECIST v 1.1.
"optimal overall remission rate" or "BOR" refers to the proportion of patients with CR or PR determined by the researcher in accordance with RECIST v 1.1.
"duration of remission" or "DOR" refers to the time from the first occurrence of recorded objective remission to disease progression or death for any reason (based on the first occurrence) as determined by the researcher in accordance with RECIST v 1.1.
"clinical benefit rate" or "CBR" refers to the proportion of patients whose disease has been determined by the researcher to be stable for at least 24 weeks or to have confirmed complete or partial remission according to RECIST v 1.1.
"Total lifetime" or "OS" refers to the time from entry into a group to death for any reason.
"time to pain deterioration (TTD)" refers to the time from randomization to the first recording of the "most severe pain" program from the brief pain scale-short version (BPI-SF) increased by ≡2 points from baseline.
"time to deterioration in body function (TTD)" refers to the time from randomization to the first recording of a decrease from baseline by ≡10 points in the European cancer research and treatment tissue quality of life core 30 questionnaire (EORTC QLQ-C30) body function scale (items 1-5).
"time to deterioration of character function (TTD)" refers to the time from randomization to the first recording of a decrease from baseline by ≡10 points in the EORTC QLQ-C30 character function table (items 6 and 7).
"Total health (GHS)/Time To Deterioration (TTD) of health-related quality of life (HRQol)" refers to the time from randomization to first recording to a decrease of ≡10 points from baseline in the EORTC QLQ-30GHS/HRQol scale (items 29 and 30).
"progression free survival" or "PFS" refers to the time from the time of entry to the first recorded date of disease progression or death (based on the first producer) determined by the researcher based on RECIST v 1.1.
"complete remission" or "CR" refers to the disappearance of all target lesions and non-target lesions and (if applicable) -normalization of tumor marker levels.
"partial remission", "PR" or "non-CR/non-PrD" refers to the continued presence of one or more non-target lesions and/or, if applicable, the maintenance of tumor marker levels above normal limits. PR may also refer to a reduction of greater than or equal to 30% in the sum of target lesion diameters, with no clear progression of CR, new lesions, and non-target lesions.
"progressive disease" or "PrD" refers to a sum of diameters of target lesions that increases by 20% or more, clear progression of non-target lesions occurs, and/or new lesions appear.
"stable disease" or "SD" refers to either insufficient shrinkage to meet the requirements of CR or PR, or insufficient tumor growth enhancement to meet the requirements of PrD.
"administration period" or "cycle" refers to a period of time that includes administration of one or more agents disclosed herein and optionally a period of time that does not include administration of one or more agents disclosed herein. For example, the total duration of one cycle may be 28 days, and include a rest period of 21 days and 7 days with administration of one or more agents. "rest period" refers to a period of time in which at least one of the agents disclosed herein is not administered. In one embodiment, the rest period refers to a period of time in which none of the agents disclosed herein are administered. In one embodiment, one cycle does not include any rest periods.
"dosing regimen" refers to a period of time comprising one or more cycles of administration of an agent described herein, wherein each cycle may comprise administration of an agent described herein in different amounts at different points in time.
"QD" refers to the administration of a compound once daily.
The graded adverse events refer to a severity grading table as established by NCI CTCAE. In one embodiment, adverse events are ranked according to the following table.
The term "detection" includes any means of detection, including direct and indirect detection.
The term "prognosis" as used herein refers to the prediction of the likelihood of death or progression of a neoplastic disease, such as cancer, due to cancer, including, for example, recurrence, metastatic spread, and drug resistance.
The term "prediction" (and grammatical variations such as other prediction lines) is used herein to refer to the likelihood that a patient will respond favorably or negatively to a drug or group of drugs. In one embodiment, the prediction is related to the degree of relief. In another embodiment, the prediction is as to whether the patient survives and/or is likely to survive after treatment, e.g., treatment with a particular therapeutic agent and/or surgical excision of the primary tumor and/or chemotherapy for a period of time without cancer recurrence. By selecting the most appropriate treatment regimen for any particular patient, the predictive methods of the invention can be used clinically to make treatment decisions. The predictive methods of the invention are valuable tools in predicting whether a patient is likely to respond favorably to a treatment regimen (e.g., a given treatment regimen), e.g., including administration of a given therapeutic agent or combination, surgical intervention, chemotherapy, etc., or whether it is likely to survive a patient for a prolonged period of time following a treatment regimen.
The term "increased resistance" to a particular therapeutic agent or treatment choice when used in accordance with the present invention refers to a reduced response to a standard dose of drug or to a standard therapeutic regimen.
Any endpoint that indicates benefit to the patient may be used to assess the "response," including but not limited to: (1) Inhibit tumor growth to some extent, including slowing or stopping growth altogether; (2) reducing the number of tumor cells; (3) reducing the tumor size; (4) Inhibit (e.g., reduce, slow or completely stop) tumor cell infiltration into adjacent surrounding organs and/or tissues; (5) Inhibit (e.g., reduce, slow or stop altogether) the transfer; (6) Enhancing an anti-tumor immune response, possibly but not necessarily resulting in tumor regression or rejection; (7) To some extent, alleviate one or more symptoms associated with a tumor; (8) increasing survival time after treatment; and/or (9) a reduction in mortality at a given point in time after treatment.
A "biomarker" is a feature that objectively measures and evaluates an indicator of normal biological processes, pathogenic processes, or pharmacological responses to therapeutic interventions. Biomarkers can be of several types: predictive, or Pharmacodynamic (PD). Predictive biomarkers predict which patients may respond or benefit from a particular therapy. The prognostic biomarker predicts the likely course of a patient's disease and may guide treatment. Pharmacodynamic biomarkers confirm drug activity and optimize dosage and administration schedule.
By analyzing a biological sample using one or more methods commonly used to establish Pharmacodynamics (PD), a "change" or "modulation" of biomarker status can be detected, including PIK3CA mutations or a set of PIK3CA mutations that occur in vitro or in vivo, comprising: (1) Sequencing the genomic DNA of the biological sample or the reverse transcription PCR product, thereby detecting one or more mutations; (2) Assessing the level of gene expression by quantifying the level of information or assessing the copy number; and (3) analyzing the protein by Immunohistochemistry (IHC), immunocytochemistry, ELISA, or mass spectrometry, thereby detecting degradation, stabilization, or post-translational modification of the protein, such as phosphorylation or ubiquitination.
"chemotherapeutic agents" are biological (macromolecular) or chemical (small molecule) compounds useful in the treatment of cancer, regardless of their mechanism of action.
The term "package insert" is used to refer to instructions that are typically included in commercial packages of therapeutic products that contain information about the indication, usage, dosage, administration, contraindications, and/or warnings that use the therapeutic products.
The phrase "pharmaceutically acceptable salt" as used herein refers to pharmaceutically acceptable organic or inorganic salts of the compounds of the present invention. Exemplary salts include, but are not limited to, sulfate, citrate, acetate, oxalate, hydrochloride, bromate, iodate, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisate, fumarate, gluconate, glucuronate, gluconate, formate, benzoate, glutamate, mesylate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate (i.e., 1' -methylene-bis- (2-hydroxy-3-naphthoic acid)) salts. The pharmaceutically acceptable salt may be directed to include another molecule, such as an acetate ion, a succinate ion, or other counterion. The counterion can be any organic or inorganic moiety that stabilizes the charge on the parent compound. Furthermore, a pharmaceutically acceptable salt may have more than one charged atom in its structure. Examples where multiple charged atoms are part of a pharmaceutically acceptable salt may have multiple counter ions. Thus, a pharmaceutically acceptable salt may have one or more charged atoms and/or one or more counter ions.
The desired pharmaceutically acceptable salts may be prepared by any suitable method available in the art. For example, the free base is treated with: if the compound of formula (I) is a base, the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treating the free base with an inorganic acid (such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, methanesulfonic acid, phosphoric acid, and the like) or with an organic acid (such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, glucopyranoside acid such as glucuronic acid or galacturonic acid, alpha-hydroxy acid such as citric acid or tartaric acid, amino acid such as aspartic acid or glutamic acid, aromatic acid such as benzoic acid or cinnamic acid, sulfonic acid such as p-toluenesulfonic acid or ethanesulfonic acid, and the like). Are generally considered suitable for use in the formation of pharmaceutically useful or acceptable salts from basic pharmaceutical compoundsThe acid is for example discussed by the following: stahl et al, camill G. (editions) Handbook of Pharmaceutical salts properties, selection and use (2002) Zurich: wiley-VCH; berge et al Journal of Pharmaceutical Sciences (1977) 66 (1) 1.19; gould, international J.of pharmaceuticals (1986) 33 201 217; anderson et al The Practice of Medicinal Chemistry (1996), academic Press, new York; remington's Pharmaceutical Sciences,18 th ed., (1995) Mack Publishing Co., easton Pa; and in The Orange Book (Food)&Drug Administration, washington, d.c., on its website). These disclosures are incorporated herein by reference.
The phrase "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the patient being treated therewith.
As used herein, the term "synergistic" refers to a therapeutic combination that is more effective than the additive effects of two or more single agents. Determination of the synergistic interaction between the compound inanolisib or a pharmaceutically acceptable salt thereof and one or more chemotherapeutic agents may be based on the results obtained from the assays described herein. Analysis of these test results can use Chou and Talalay combination methods anddose-effect analysis of the software to obtain a combination index (Chou and Talalay,1984,Adv.Enzyme Regul.22:27-55). The combinations provided by the present invention have been evaluated in several assay systems and data can be analyzed using standard procedures for quantifying synergy, addition, and antagonism between anticancer agents as described by Chou and Talalay in "New Avenues in Developmental Cancer Chemotherapy," Academic Press,1987, chapter 2. A combination index value of less than 0.8 indicates synergy, a value of greater than 1.2 indicates antagonism, and a value between 0.8 and 1.2 indicates additive effects. Combination therapy may provide "synergy" and prove "synergistic", i.e., an effect achieved when the active ingredients are used together that is greater than the sum of the effects produced by the compounds alone. When the effective components are as follows, the synergistic effect can be achieved: (1) Co-formulation Administered in a combined unit dosage formulation or delivered simultaneously; (2) delivered alternately or in parallel in separate formulations; or (3) by some other scheme. When delivered in alternating therapy, a synergistic effect may be obtained when the compounds are administered or delivered sequentially, for example in different syringes or in separate pills or tablets. Generally, during alternating therapy, the effective dosages of each active ingredient are administered sequentially, i.e., serially, while in combination therapy, the effective dosages of two or more active ingredients are administered together. The combined effect was evaluated using the BLISS independence model and the Highest Single Agent (HSA) model (Leh ar et al, 2007,Molecular Systems Biology 3:80). BLISS score quantifies the degree of potentiation of a single dose, whereas BLISS score>A0 indicates that it is larger than a simple addition. HSA fraction>0 means that the combined effect is greater than the maximum of the single agent response at the corresponding concentration.
Clinical compounds
inavelisib (also known as GDC-0077):
inavelisib is a potent, orally bioavailable, clinically-stage class I PI3K alpha isoform (PI 3K alpha) selective inhibitor with > 300-fold less potent biochemical inhibition of other class I PI3K beta, delta, and gamma isoforms, and has increased potency to tumor cells bearing mutant PI3 ks as compared to wild-type (WT) PI3K cells (Braun, m.et al, "Discovery of GDC-0077:A highly selective inhibitor of PI3K-alpha that induces degradation of mutant-p110 alpha protein" Abstracts of Papers,254th ACS National Meeting&Exposition,Washington,DC,USA,August 20-24,2017, medi-22; garland, k.et al, "Discovery of novel class of alpha selective PI K inhibitors" Abstracts of Papers,254th ACS National Meeting&Exposition,Washington,DC,USA,August 20-24,2017, medi-103; hojr.et al, "GDC-0077is a selective PI3K alpha inhibitor that demonstrates robust efficacy in PIK3CA mutant breast cancer models as a single agent and in combination with standard of care therapies"2017San Antonio Breast Cancer Symposium,Dec.5-9 2017,San Antonio,TX,Abstract Publication Number:PD4-14; edgar, k.et al, "Preclinical characterization of GDC-0077,aspecific PI3K alpha inhibitor in early clinical development"Cancer Research77 (13 journals 156July 2017).
Inavaoliib, CAS registry number 2060571-02-8, gen-Tek, U.S. 9650393; named (S) -2- ((2- ((S) -4- (difluoromethyl) -2-dioxooxazolidin-3-yl) -5, 6-dihydrobenzo [ f ] imidazo [1,2-d ] [1,4] oxazin-9-yl) amino) propanamide having the following structure:
inavaoliib is also known as GDC-0077, RG6114, RO7113755, or the chemical name (2S) -2- [ [2- [ (4S) -4- (difluoromethyl) -2-oxo-3-oxazolidinyl ] -5, 6-dihydroimidazo [1,2-d ] [1,4] benzooxazepin-9-yl ] amino ] propionamide.
GDC-0077 inhibits membrane-bound 4, 5-phosphatidylinositol diphosphate (PIP) by binding to the ATP binding site of PI3K 2 ) Phosphorylation to 3,4, 5-phosphatidylinositol triphosphate (PIP) 3 ) To exert its activity. Inhibiting PIP 2 To PIP 3 Reduces downstream activation of AKT and pS6, resulting in reduced cell proliferation, metabolism, and angiogenesis. Non-clinical studies showed that GDC-0077 specifically degraded mutant p110α, inhibited proliferation and induced apoptosis of PIK3CA mutant breast cancer cell lines, inhibited tumor growth in human breast cancer xenograft models containing PIK3CA mutations, and reduced downstream PI3K pathway markers including phosphorylated form of protein kinase B (pAKT), PRAS40 phosphorylated at threonine 246 (pPRAS 40), and S6RP phosphorylated at serine 235/236 (pS 6 RP).
Trastuzumab:
trastuzumab (CAS 180288-69-1),huMAb4D5-8, rhuMAb HER2, geneva) is a recombinant DNA derived IgG1 kappa monoclonal antibody, a humanized version of the murine anti-HER 2 antibody (4D 5), based onThe extracellular domain of HER2 binds selectively with high affinity (Kd=5 nM) in assays of cells (US 5677171;US 5821337;US 6054297;US 6165464;US 6339142;US 6407213;US 6639055;US 6719971;US 6800738;US 7074404;Coussens et al (1985) Science 230:1132-9; slamon et al (1989) Science 244:707-12; slamon et al (2001) New Engl. J. Med. 344:783-792). Trastuzumab has been shown to inhibit proliferation of HER2 over-expressed human tumor cells both in vitro assays and in animals (Hudziak et al (1989) Mol Cell Biol 9:1165-72; lewis et al (1993) Cancer Immunol Immunother;37:255-63; basega et al (1998) Cancer Res.58:2825-2831). Trastuzumab is a mediator of antibody-dependent cytotoxicity ADCC (Lewis et al (1993) Cancer Immunol Immunother 37 (4): 255-263; hotaling et al (1996) [ abstract)]Proc.annu Meeting Am Assoc Cancer Res;37:471; pegram MD, et al (1997) [ abstreact ] ]Proc Am Assoc Cancer Res;38:602; sliwkowski et al (1999) Seminars in Oncology (4), suppl 12:60-70; yarden Y. And Sliwkowski, M. (2001) Nature Reviews: molecular Cell Biology, macmillan Magazines, ltd., vol.2: 127-137).
(trastuzumab) was approved in 1998 for treatment of patients with metastatic breast cancer that had been over-expressed in HER2 with past anti-cancer therapy (Baselga et al, (1996) J.Clin.Oncol.14:737-744), after which it has been used in more than 300,000 patients (Slamon DJ, et al N Engl J Med 2001;344:783-92; vogel CL, et al J Clin Oncol 2002;20:719-26; marty M, et al J Clin Oncol 2005;23:4265-74; romond EH, et al T N Engl J Med 2005; 1673-84; piccrart-Gebhart MJ, et al NEngl J Med 2005;353:1659-72; slamon D, et al [ abs reacting ]]Breast Cancer Res Treat 2006,100 (Suppl 1): 52). In 2006, FDA approval ++>(trastuzumab, geneTex) as a treatment regimen comprising doxorubicin (doxorubicin), cyclophosphamide and paclitaxelAnd part, is used for adjuvant therapy of HER2 positive and lymph node positive breast cancer patients.
HERCEPTIN HYLECTA TM (trastuzumab and hyaluronidase-oysk) is a combination of trastuzumab and recombinant human hyaluronidase (an endoglycosidase, a tissue permeability regulator administered by subcutaneous fluid), which has been approved by the us FDA for the treatment of HER2 overexpressing breast cancer.
Pertuzumab:
pertuzumab (also known as recombinant humanized monoclonal antibody 2C4, rhuMAb 2C4,The gene tek corporation, san francisco, represents the first of a new class of agents known as HER Dimerization Inhibitors (HDI) that function to inhibit the ability of HER2 to form active heterodimers or homodimers with other HER receptors (e.g., EGFR/HER1, HER2, HER3, and HER 4). See, e.g., harari and Yarden Oncogene 19:6102-14 (2000); yarden and Sliwkowski. Nat Rev Mol Cell Biol 2:127-37 (2001); sliwkowski Nat Struct Biol 10:158-9 (2003); cho et al Nature 421:756-60 (2003); and Malik et al Pro Am Soc Cancer Res 44:176-7 (2003).
(pertuzumab) was first approved in 2012 for the treatment of advanced or later stage (metastatic) HER2 positive breast cancer patients. The U.S. food and drug administration accelerated approval +.f. at 30, 9, 2013>(pertuzumab) as part of a complete treatment regimen prior to surgery (neoadjuvant treatment) for patients with early stage breast cancer (EBC).For the first time FDA approved drugs for new adjuvant treatment of breast cancer were obtained.
(pertuzumab, trastuzumab and hyaluronidase-zzxf) contains pertuzumab and a fixed dose combination of trastuzumab and hyaluronidase for subcutaneous injection, and has been approved by the us FDA for the treatment of early and metastatic HER2 positive breast cancer in 2020. PHESGO can be administered by subcutaneous (SC; subcutaneous) injection in combination with Intravenous (IV) chemotherapy.
Pertuzumab inhibits ligand-initiated intracellular signaling through the primary signaling pathway (including PI 3K), which can lead to cell growth arrest and apoptosis. Furthermore, both trastuzumab and pertuzumab mediate antibody-dependent cellular cytotoxicity (ADCC). Overall, the combination of pertuzumab and trastuzumab was well tolerated without a significant increase in left ventricular contractile dysfunction.
Fulvestrant:
fulvestrant is an ER antagonist and is effective in the treatment of post-menopausal hr+ breast cancer patients with relatively preferred tolerance. The expected toxicity of GDC-0077 and fulvestrant did not overlap. It is important to test the use of GDC-0077 in combination with both letrozole and fulvestrant because these endocrine therapies have different mechanisms of action, different PK properties and different drug-to-drug interaction (DDI) possibilities with inavillisib.
Fulvestrant @AstraZeneca, CAS Reg.No.129453-61-8) has acquired FDA approval for the treatment of hormone receptor positive (HR+) metastatic breast cancer in postmenopausal women following antiestrogen treatment for disease progression (Kansra (2005) Mol Cell Endocrinol 239 (1-2): 27-36; flemming et al, (2009) Breast Cancer Res treat. 115 (2) 255-68; valachis et al, (2010) Crit Rev Oncol Hematol. Mar;73 (3):220-7). Fulvestrant is an Estrogen Receptor (ER) antagonist without agonist action that acts by both down-regulation and degradation of the estrogen receptor (Croxital (2011) Drugs 71 (3): 363-380). Fulvestrant is also a selective estrogen receptor down-regulator (SERD).
Fulvestrant is designated (7α,17β) -7- {9- [ (4, 5-pentafluoropentyl) sulfinyl ] nonyl } estra-1, 3,5 (10) -triene-3, 17-diol and has the following structure:
fulvestrant belongs to a reversible steroid ER antagonist that competes directly for ER binding with female sex hormone and lacks the partial agonist properties of tamoxifen. After binding to ER, it blocks estrogen signaling and increases ER protein degradation. Fulvestrant has an affinity for ER that is about 100 times greater than that of tamoxifen (Howell et al, (2000) Cancer 89:817-25). Fulvestrant (250 mg once per month) was FDA approved in 2002 and EMA approved in 2004 for use in treating HR positive MBC in postmenopausal women for disease progression following antiestrogenic therapy. In a multicenter phase III study, fulvestrant was found to be at least equivalent to anastrozole (a non-steroidal anti-inflammatory drug AI) in the second line treatment setting (Howell et al, (2002) J Clin Oncol 20:3396-3403; osborne CK et al, (2002) J Clin Oncol 20:3386-95). Fulvestrant is also as effective as tamoxifen in the first line treatment of advanced breast cancer (Howell et al, (2004) JClin Oncol 22:1605-1613) and shows a level of activity in patients with post-AI metastatic disease conditions similar to the nonsteroidal anti-inflammatory drug AI exemestane (Chia et al, (2008) J Clin Oncol 26:1664-1670). High doses of fulvestrant (500 mg once a month) have been demonstrated to be at least as effective as anastrozole in terms of Clinical Benefit Rate (CBR) and overall remission rate, and significantly correlated with the time of progression preference for first line treatment in women with advanced HR positive breast cancer (Robertson et al, (2009) J Clin Oncol 27:4530-4535). High doses of fulvestrant have recently been shown to have excellent disease Progression Free Survival (PFS) in women with ER-positive advanced breast cancer with 500mg treatment relative to patients treated with 250mg (Di Leo et al, (2010) J Clin Oncol 28:4594-4600). In these studies fulvestrant (250 mg and 500 mg) was well tolerated and produced less female effect compared to tamoxifen and less joint pain compared to AI anastrozole (Osborne et al, (2002) JClin Oncol 20:3386-3395). These results led to approval of 500mg fulvestrant once a month as the recommended dose currently approved in the united states and the european union (2010) for postmenopausal women with spread of disease after AI treatment. These studies indicate that fulvestrant is an important treatment choice for patients with advanced breast cancer and is therefore considered an appropriate control therapy for this study.
Letrozole:
letrozole is effective in treating post-menopausal hr+ breast cancer patients with relatively good tolerability. inanolisib does not overlap with the expected toxicity of letrozole. Letrozole @Novartis pharm.) is an oral non-steroidal aromatase inhibitor for the treatment of post-operative hormone-responsive breast cancer (Bhatnagar et al, (1990) j. Steroid biochem. And mol. Biol 37:1021; lipton et al, (1995) Cancer75:2132; goss, p.e. and Smith, r.e. (2002) Expert rev. Anticancer ter. 2:249-260; lang et al, (1993) The Journalof Steroid biochem. And mol. Biol.44 (4-6): 421-8; EP 236940; US 4978672). />FDA approval has been obtained for the treatment of hormone receptor positive (hr+) or localized or metastatic breast cancer with unknown receptor status in postmenopausal women.
Letrozole is designated as 4,4' - ((1H-1, 2, 4-triazol-1-yl) methylene) dibenznitrile (CAS Reg. No. 112809-51-5) and has the structure:
paclitaxel:
paclitaxel is a chemotherapeutic drug approved for the treatment of various types of cancer (e.g., ovarian, breast, lung, etc.), as monotherapy or in combination with other anticancer agents. Paclitaxel is designated 5 beta, 20-epoxy-1, 2 alpha, 4,7 beta, 10 beta, 13 alpha-hexahydroxytaxane-11-en-9-one 4, 10-diacetate 2-benzoate 13- (2 r,3 s) -N-benzoyl-3-phenylisoserine and has the following structure:
Combination therapy
In human epidermal growth factor receptor 2 positive (her2+) breast cancers, dysregulation of the PI3K/AKT/mTOR pathway, activating mutations and other distorted forms, has been identified as a possible mechanism for developing resistance to HER 2-targeted therapies. Thus, continued inhibition of the HER2 pathway and co-targeting of the PI3K/AKT/mTOR pathway may restore sensitivity to HER 2-targeted therapies. The addition of PI3K inhibitors to trastuzumab and pertuzumab may improve the outcome of PIK3CA mutant her2+ breast cancer patients.
Provided herein are combination or combination therapies comprising inanolisib (GDC-0077) for HER 2-targeted therapy. In some embodiments, the HER 2-targeted therapy is trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab. In one embodiment, the combination or combination therapy comprises inanolisib, trastuzumab, and pertuzumab. In one embodiment, the combination or combination therapy further comprises hyaluronidase. In one embodiment, the combination or combination therapy comprises inavilisib, trastuzumab and hyaluronidase-oysk, and pertuzumab. In one embodiment, the combination or combination therapy comprises inaviolisib, pertuzumab, trastuzumab, and hyaluronidase-zzxf.
The combination or combination therapy as described herein may be provided as a kit comprising one or more agents for administration. In one embodiment, the kit comprises inanolisib and trastuzumab. In one embodiment, the kit comprises inavilisib, trastuzumab and hyaluronidase-oysk. In one embodiment, the kit comprises inanolisib and pertuzumab. In another embodiment, the kit comprises inavillisib, trastuzumab and pertuzumab. In another embodiment, the kit comprises inavillisib and a fixed dose combination of trastuzumab and pertuzumab. In another embodiment, the kit comprises inavilisib, pertuzumab, trastuzumab, and hyaluronidase-zzxf. In one embodiment, the agents of the combination or combination therapies described herein are supplied in a kit in a form ready for administration. The kits disclosed herein can include instructions for use, such as package inserts. In one embodiment, the instructions for use are one portion of each reagent in the package insert-kit.
Further provided are kits for practicing the methods detailed herein, comprising a pharmaceutical composition or combination therapy as described herein and instructions for treating breast cancer.
The kit typically includes suitable packaging. The kit may comprise one or more containers comprising any pharmaceutical composition as described herein. Each component (if there is more than one component) may be packaged in a separate container or some components may be combined in one container where cross-reactivity and shelf life are allowed. One or more components of the kit may be sterile and/or may be contained within a sterile package.
Method
HER2 overexpression is an important prognostic and predictive biomarker for metastatic breast cancer (Pauletti et al 2000). In phase III studies of trastuzumab plus docetaxel with or without pertuzumab in 1l her2+mbc patients, higher than median (high) HER2 protein expression as measured by immunohistochemistry and higher than median (high) HER2 mRNA expression as measured by qRT-PCR (quantitative reverse transcription polymerase chain reaction) correlated significantly with better prognosis in this patient population (HR 0.83[ p=0.05 ] and HR 0.77[ p=0.008 ], respectively) (Baselga, et al 2014). Frequent PIK3CA mutations were also observed in patients with HER2 expression, and there was a difference in prevalence of PIK3CA mutations. Among patients who observed PIK3CA mutations, IHC showed that the number of patients whose tumors had focal, heterogeneous HER2 expression (42%) was almost twice the number of patients whose tumors had robust homogeneous HER2 expression (24%) (Perez et al 2019). Thus, it would be beneficial to treat her2+ breast cancer using a combination of a mutant PI3K alpha inhibitor and HER2 targeted therapy.
All PI3K alpha inhibitors are not identical. inanolisib (GDC-0077) is more advantageous than another clinically relevant PI3K alpha inhibitor, apicalist (also known as BYL 719) in inhibiting the growth of her2+pik3ca mutant cancer cells. In one study comparing the efficacy of inavilisib and apilimbus, the results showed a significant difference between the sensitivity of inavilisib and apilimbus in HER2 expanded cell lines (about 20-fold difference between mean IC50 values) and HER2 negative cell lines (6-fold difference between the two inhibitors). Regardless of HER2 status, both inhibitors did not differentiate in PIK3CA-WT cell lines.
Provided herein are methods of treating HER2 positive cancers. In one embodiment, the method comprises treating HER2 positive breast cancer in a patient suffering from locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer by administering to the patient a combination therapy comprising inavillisib and HER2 targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab).
Also provided are methods of treating locally advanced or metastatic PIK3CA mutated (or PIK3CA mutated) HER2 positive breast cancer in a patient, comprising administering a therapeutically effective amount of inanolisib (GDC-0077) or a pharmaceutically acceptable salt thereof and HER2 targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab). In some embodiments, the HER 2-targeted therapy comprises trastuzumab. In some embodiments, the HER2 targeted therapy comprises trastuzumab and hyaluronidase-oysk. In some embodiments, the HER2 targeted therapy comprises pertuzumab. In some embodiments, the HER2 targeted therapy comprises trastuzumab and pertuzumab. In some embodiments, HER 2-targeted therapy comprises a fixed dose combination of trastuzumab and pertuzumab. In some embodiments, HER2 targeted therapies comprise pertuzumab, trastuzumab, and hyaluronidase-zzxf. In some embodiments, the method further comprises administering paclitaxel.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising: inavilisib, trastuzumab and pertuzumab; wherein the combination therapy is administered over a 21 day period. In some embodiments, the combination therapy further comprises paclitaxel.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. QDs were administered inavilisib on days 1 to 21 of the first 21 day cycle;
b. trastuzumab was administered on day 1 of the first 21-day cycle; and
c. pertuzumab was administered on day 1 of the first 21 day cycle.
In some of these embodiments, the method further comprises one or more additional 21-day cycles comprising:
a. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
b. trastuzumab was administered on day 1 of each additional 21-day cycle; and
c. Pertuzumab was administered on day 1 of each additional 21-day cycle.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 2 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle; and
d. pertuzumab was administered on day 2 of the first 21 day cycle.
In some of these embodiments, the method further comprises one or more additional 21-day periods (e.g., one, two, three, four, or five additional 21-day periods), the one or more additional 21-day periods comprising:
a. paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
b. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
c. trastuzumab was administered on day 1 of each additional 21-day cycle; and
d. pertuzumab was administered on day 1 of each additional 21-day cycle.
Depending on the Dose Limiting Toxicity (DLT) observed in the patient, inavelisib may be administered on a 6/1 or 5/2 dosing regimen. inanolisib is administered for 6 days, 1 day at rest, or 5 days, 2 days at rest per week.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 2-7, 9-14 and 16 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle; and
d. pertuzumab was administered on day 2 of the first 21 day cycle.
In some of these embodiments, the method further comprises one or more additional 21-day periods (e.g., one, two, three, four, or five additional 21-day periods), the one or more additional 21-day periods comprising:
a. paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
b. inavelisib was administered on days 2-7, 9-14 and 16 to 21 of each additional 21 day cycle;
c. Trastuzumab was administered on day 1 of each additional 21-day cycle; and
d. pertuzumab was administered on day 1 of each additional 21-day cycle.
In some embodiments, there is provided a method of treating HER2 positive breast cancer in a patient having HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutations, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 3-7, 10-14 and 17 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle; and
d. pertuzumab was administered on day 2 of the first 21 day cycle.
In some of these embodiments, the method further comprises one or more additional 21-day periods (e.g., one, two, three, four, or five additional 21-day periods), the one or more additional 21-day periods comprising:
a. paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
b. inavelisib was administered on days 3-7, 10-14 and 17 to 21 of each additional 21 day cycle;
c. Trastuzumab was administered on day 1 of each additional 21-day cycle; and
d. pertuzumab was administered on day 1 of each additional 21-day cycle.
After receiving up to six cycles of combination therapy (induction therapy) comprising paclitaxel, inanolisib, pertuzumab, and trastuzumab, the patient can continue to receive additional cycles of combination therapy (maintenance therapy) comprising inanolisib, pertuzumab, and trastuzumab (without paclitaxel) until the disease progresses or unacceptable associated toxicity occurs. During maintenance therapy, HR positive/HER 2 positive metastatic breast cancer patients may be treated with endocrine therapy (e.g., letrozole or fulvestrant).
In some of these embodiments, the method comprises administering to the patient an induction therapy comprising paclitaxel, inanolisib, pertuzumab, and trastuzumab in a regimen comprising the first 21-day period detailed herein and up to five additional 21-day periods detailed herein, further comprising a maintenance therapy comprising inanolisib, pertuzumab, and trastuzumab (without paclitaxel), in a regimen comprising:
a. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
b. Trastuzumab was administered on day 1 of each additional 21-day cycle; and
c. pertuzumab was administered on day 1 of each additional 21-day cycle.
In one embodiment, the method comprises a combination therapy comprising inanolisib and HER 2-targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab). In one embodiment, the method comprises a combination therapy comprising inavillisib, trastuzumab and pertuzumab administered according to a dosing regimen described herein. In one embodiment, the method comprises a combination therapy comprising paclitaxel, inanolisib, trastuzumab, and pertuzumab administered according to a dosing regimen described herein.
In yet another aspect, a method of inhibiting tumor growth or producing/increasing tumor regression in a patient suffering from locally advanced or metastatic PIK3CA mutant HER2 positive breast cancer is provided, the method comprising administering a combination therapy to the patient according to the methods detailed herein.
The agents disclosed herein may be administered according to a package insert. In one embodiment of the methods disclosed herein, these agents can be administered in an effective amount as disclosed herein. In some embodiments, trastuzumab and/or pertuzumab, if applicable, is administered at its approved dose by an approved route of administration.
The agents in the combination therapies detailed herein may be administered simultaneously or sequentially. Two of the three agents of the combination therapy may be administered simultaneously, while the third agent may be administered before or after. For example, in one embodiment of the methods described herein, administration of inanolisib occurs prior to administration of another dose (e.g., pertuzumab or trastuzumab). In one embodiment of the methods described herein, inavelisib is administered first, then pertuzumab is administered, and then trastuzumab is administered. In another embodiment, inavilisib is administered before or after a fixed dose combination of pertuzumab and trastuzumab (e.g., a fixed dose combination of pertuzumab, trastuzumab, and hyaluronidase-zzxf). In some embodiments, paclitaxel is administered prior to the administration of the triple combination therapy in any order detailed herein.
In some embodiments, inanolisib is administered in an amount of 3, 6, or 9mg, for example, in one or more oral tablets. In some embodiments, inanolisib is administered orally at a daily dose of 9 mg. In some of these embodiments, inanolisib is administered in an amount of 9mg, for example, in an oral tablet. In some embodiments, inanolisib is administered orally at a daily dose of 6mg, e.g., in one or more oral tablets.
In some embodiments, trastuzumab is administered by Intravenous (IV) infusion. In some embodiments, trastuzumab is administered at a loading dose of 8mg/kg during the first 21-day period. In some embodiments, trastuzumab is administered at a dose of 6mg/kg in each additional cycle. In some embodiments, trastuzumab is administered by Subcutaneous (SC) injection. In some embodiments, trastuzumab is administered with hyaluronidase-oysk by Subcutaneous (SC) injection.
In some embodiments, pertuzumab is administered by Intravenous (IV) infusion. In some embodiments, pertuzumab is administered at a loading dose of 840mg during the first 21-day period. In some embodiments, pertuzumab is administered at a dose of 840mg at each additional cycle. In some embodiments, pertuzumab is administered by Subcutaneous (SC) injection. In some embodiments, pertuzumab is administered by Subcutaneous (SC) injection with trastuzumab and hyaluronidase-zzxf.
In some embodiments, paclitaxel is administered by Intravenous (IV) infusion. In some embodiments, paclitaxel is at 80g/m 2 Is administered in a weekly dose. In some embodiments, paclitaxel is infused intravenously at 80mg/m 2 Is administered weekly.
In some embodiments, inanolisib is administered in an amount of 9mg, for example, in an oral tablet. In some embodiments, trastuzumab is administered by Intravenous (IV) infusion at a load dose of 8mg/kg during the first 21-day period and at a dose of 6mg/kg during each additional 21-day period. In some embodiments, pertuzumab is administered by IV infusion at a loading dose of 840mg during the first 21 day period and at a dose of 420mg during each additional 21 day period.
In one embodiment, the methods disclosed herein comprise administering the combination therapies disclosed herein according to a dosing regimen comprising a 21-day period. In another embodiment, the methods disclosed herein comprise a combination therapy disclosed herein administered according to a dosing regimen comprising a first 21-day cycle followed by an additional 21-day cycle. In another embodiment, the methods described herein comprise a combination therapy described herein administered according to a dosing regimen comprising a first 21-day period followed by 2 to 30 additional 21-day periods, or until disease progression or unacceptable toxicity occurs. In yet another embodiment, the methods described herein comprise the induction therapy described herein administered according to a dosing regimen comprising a first 21-day period followed by up to five additional 21-day periods, or until disease progression or unacceptable toxicity occurs. In another embodiment, the methods described herein comprise an induction therapy described herein administered according to a dosing regimen comprising a first 21-day cycle followed by up to five additional 21-day cycles, and a maintenance therapy described herein administered according to a dosing regimen comprising one or more additional 21-day cycles (e.g., up to 30 additional 21-day cycles), or until the disease progresses or unacceptable toxicity occurs.
Efficacy of the combination is measured in terms of Progression Free Survival (PFS), total survival (OS), objective Remission Rate (ORR), and other relevant clinical outcomes.
In some embodiments, the patient has PIK3CA mutant HER2 positive locally advanced or metastatic breast cancer. In some embodiments, the patient is a female patient with a histologically recorded locally advanced or metastatic PIK3CA mutant her2+ breast cancer. The patient's hormone receptor status may be positive or negative. In some embodiments, the patient has HER2 positive breast cancer with hormone receptor positive (hr+) locally advanced or metastatic PIK3CA mutations. In some embodiments, the patient has HER2 positive breast cancer with a hormone receptor negative (HR-) locally advanced or metastatic PIK3CA mutation.
In some embodiments, the patient has a mutant PIK3CA having a mutation at one or more of positions 88, 106, 111, 118, 345, 420, 453, 542, 545, 546, 1043, 1047, and 1049. In some embodiments, the patient has a mutant PIK3CA having a mutation in one or more of H1047, E545, E542, Q546, N345, C420, M1043, G1049, E453, K111, G106, G118, and R88. In some embodiments, the patient has a mutant PIK3CA comprising one or more mutations selected from the group consisting of: H1047D/I/L/N/P/Q/R/T/Y, E545A/D/G/K/L/Q/R/V, E542A/D/G/K/Q/R/V, Q546E/H/K/L/P/R, N345D/H/I/K/S/T/Y, C420/3552R, M1043I/T/V, G1049A/C/D/R/S, E453A/D/G/K/Q/V, K111N/R/E, G A/D/R/S/V, G118D, and R88Q. In some embodiments, the patient has a mutant PIK3CA comprising one or more mutations selected from the group consisting of: E542K, E545K, Q546R, H1047L and H1047R. In some embodiments, the patient has breast cancer that expresses a PIK3CA mutation selected from the group consisting of: H1047D/I/L/N/P/Q/R/T/Y, E545A/D/G/K/L/Q/R/V, E542A/D/G/K/Q/R/V, Q546E/H/K/L/P/R, N345D/H/I/K/S/T/Y, C420/3552R, M1043I/T/V, G1049A/C/D/R/S, E453A/D/G/K/Q/V, K111N/R/E, G A/D/R/S/V, G118D, and R88Q. In some embodiments, the patient has breast cancer expressing a PIK3CA mutant selected from the group consisting of: H1047R/Y/L, E542K, E545K/D/G/A, Q K/R/E/L, N345K, C420R, G1049R, R88Q and M1043I. In some embodiments, the patient has breast cancer that expresses a PIK3CA mutation selected from the group consisting of: E542K, E545K, Q546R, H1047L and H1047R.
In some embodiments, the patient has a mutant PIK3CA comprising a mutation selected from the group consisting of: E542K, E545K, Q546R, H1047L and H1047R, and a second mutation (e.g., a second mutation selected from E453Q/K, E726K and M1043L/I). In some embodiments, the patient has breast cancer that expresses a PIK3CA mutation that expresses a double mutation selected from the group consisting of: e542K+E453Q/K, E542K+E726K, E542K+M1043L/I; e545K+E453Q/K, E545K+E726K, E545K+M1043L/I; h1047r+e453Q/K, and h1047r+e726K.
PIK3CA mutant tumor status can be assessed by blood concentration testing or local testing of blood or tumor tissue. In some embodiments, the focused test for qualifying PIK3CA mutations is a foundation one fluid clinical trial assay (Liquid Clinical Trial Assay) conducted at basic medicine company (Foundation Medicine, inc.). In some embodiments, local testing of blood or tumor tissue is performed in CLIA-certified or equivalent laboratories using PCR-or NGS-based assays that are pre-approved by sponsors.
The HER2 status of the tumor can be assessed by HER2 protein over-expression and/or HER2 gene amplification in the tumor sample using known methods (e.g., FDA approved assays for detecting HER2 protein over-expression and HER2 gene amplification), preferably by a laboratory with demonstrated capability using FDA approved assays specific for breast cancer. HER2+ is defined as (i) HER2 IHC score of 3+; (ii) HER2 IHC score of 2+ with a dual probe ISH HER2/CEP17 ratio of > 2.0 (preferred); (iii) Fluorescence, chromogenic or silver ISH tests indicate the presence of HER2 gene amplification; or (iv) her2+ according to local clinical guidelines.
In some embodiments, the patient is a patient with a histologically recorded locally advanced or metastatic PIK3CA mutant her2+ breastFemale patients with cancer. In some embodiments, the patient has a HER2 IHC score of 3 or more. In some embodiments, the patient has a HER2 IHC score of 2 or more with a dual probe ISH HER2/CEP17 ratio of no less than 2.0 (preferred). In some embodiments, the patient has a fluorescent, chromogenic, or silver ISH test result that indicates the presence of HER2 gene amplification. In some embodiments, the patient is her2+ according to local clinical guidelines. In some embodiments, the patient has a fasting blood glucose level equal to or less than 140 mg/dL. In some embodiments, the patient has less than 7% glycosylated hemoglobin (HbA) 1c ) Horizontal. In some embodiments, the patient has a Left Ventricular Ejection Fraction (LVEF) of ≡50%. LVEF may be determined by Echocardiography (ECHO) (preferred) or multiple gated acquisition (MUGA) scans. In some embodiments, the patient has received a prior HER 2-targeted therapy (e.g., trastuzumab and/or pertuzumab) and has not discontinued the prior HER 2-targeted therapy (e.g., trastuzumab and/or pertuzumab) because toxicity was assessed as being associated with the prior HER 2-targeted therapy (e.g., trastuzumab and/or pertuzumab).
Patients may have hr+ or HR-breast cancer. Hr+ is defined as the presence of ER expression in ≡1% of cells, or hr+ according to local clinical guidelines. HR-is defined as the presence of ER expression in <1% of cells, or HR-according to local clinical guidelines. Hr+ breast cancer patients may be treated with endocrine therapy (e.g., letrozole or fulvestrant) and, at the discretion of the investigator, pre-menopausal or perimenopausal patients may also be treated with LHRH agonist therapy.
In some embodiments, the patient has hr+ locally advanced or metastatic PIK3CA mutant her2+ breast cancer, and the method of treatment comprises administering to the patient a combination therapy comprising inanolisib, trastuzumab, pertuzumab, and endocrine therapy (e.g., letrozole or fulvestrant).
In some embodiments, a method for treating hr+ locally advanced or metastatic her2+ breast cancer in a patient is provided, the method comprising administering to the patient a therapeutically effective amount of PIK3CA mutant inanolisib or a pharmaceutically acceptable salt thereof, trastuzumab, pertuzumab, and an endocrine therapy treatment (e.g., letrozole or fulvestrant). In some embodiments, the endocrine therapy is letrozole administered according to its approved label, e.g., daily in oral tablets at a dose of 2.5 mg. In some embodiments, the endocrine therapy is fulvestrant administered according to its approved label, e.g., administered at a dose of 500mg about once every four weeks by Intramuscular (IM) infusion.
In one embodiment of the methods described herein, the patient was treated with one or more cancer therapies prior to administration of the combination therapies described herein. In one embodiment of the methods described herein, the prior therapy comprises HER 2-targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab). In another embodiment, the patient described herein has not been previously treated with HER 2-targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab).
In one embodiment of the methods described herein, the patient has a breast cancer described herein that is resistant to one or more cancer therapies (e.g., HER 2-targeted therapies, such as trastuzumab or pertuzumab). In one embodiment of the methods described herein, the resistance to cancer therapy comprises cancer recurrence or refractory cancer. Recurrence may refer to the recurrence of cancer at the original or new site after treatment. In one embodiment of the methods described herein, the resistance to a cancer therapy comprises progression of the cancer during treatment with an anti-cancer therapy. In some embodiments of the methods described herein, resistance to cancer therapy includes cancer that does not produce remission for treatment. The cancer may be resistant at the beginning of the treatment or it may become resistant during the course of the treatment. In some embodiments of the methods described herein, the cancer is in an early or late stage.
Coadministration of inanolisib with HER2 targeted therapies (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab) can prevent or delay the development of resistance of a tumor (e.g., breast cancer) to HER2 targeted therapies (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab). Thus, there is provided a method of preventing or delaying the development of resistance of a tumor (e.g., breast cancer) to a therapy containing HER 2-targeted therapy (e.g., trastuzumab, pertuzumab, or a combination of trastuzumab and pertuzumab), the method comprising administering a combination therapy as detailed herein. In some embodiments, a method of preventing or delaying the development of resistance of a tumor (e.g., breast cancer) to a therapy containing trastuzumab and/or pertuzumab is provided, the method comprising administering a combination therapy comprising trastuzumab and/or pertuzumab or a combination therapy comprising inanolisib, trastuzumab and pertuzumab. In some embodiments, the combination therapy is administered according to any of the methods as detailed herein.
Biomarkers and methods for their use
Breast cancer is a heterogeneous disease with a number of distinct subtypes defined by molecular signatures and a wide variety of mutation profiles. In one embodiment, patients may be tested for PIK3CA/AKT1/PTEN change status. In one embodiment, the patient disclosed herein may be tested for one or more of phosphatase and tensin homolog (PTEN) mutations, loss of PTEN expression, phosphatidylinositol-4, 5-biphosphate 3-kinase catalytic subunit alpha (PIK 3 CA) mutations, protein kinase B alpha (AKT 1) mutations, or combinations thereof. In one embodiment, the loss of PTEN expression is hemizygous or homozygous. In another embodiment, additional biomarkers of a sample of a patient disclosed herein can be evaluated to assess factors that may be correlated with the safety and efficacy of the study treatment.
In one embodiment of the methods described herein, NGS, whole Genome Sequencing (WGS), other methods, or combinations thereof, may be used for DNA obtained from blood samples and tumor tissue from the patients described herein. Such samples can be analyzed to identify germ line (e.g., BRCA 1/2) and somatic alterations that predict remission of the study drug, correlate with progression to a more severe disease state, correlate with acquired resistance to the study drug, or increase knowledge and understanding of disease biology. In another embodiment of the methods disclosed herein, a patient disclosed herein may have a cancer characterized by activation of PI3K/Akt signals, such as activating mutations in PIK3CA or Akt1, and by alterations in PTEN, such as those provided herein. In another embodiment, the altered tumor status of PIK3CA/AKT1/PTEN will be determined using NGS assays (e.g., medical foundation company (Foundation Medicine, inc. [ FMI ])). Status of PIK3CA/AKT1/PTEN changes in the archiving tissue and remission measurements may be performed continuously. Expression of a biomarker provided herein (e.g., PTEN) can be measured using techniques known in the art such as, for example, immunohistochemistry (IHC).
Circulating tumor DNA (ctDNA) can be detected in the blood of Cancer patients with epithelial cancers and can have diagnostic and therapeutic implications (Schwarzenbach et al Nat Rev Cancer 2011; 11:426-437). For example, tumor cells in the mutated state can be obtained by isolation of ctDNA (Maheswaran S, et al N Engl J Med 2008; 359:366-77), and ctDNA has been used to monitor therapeutic effectiveness in melanoma (Shinozaki M, et al Clin Cancer Res 2007; 13:2068-74). Blood samples from the patients disclosed herein may be collected at the time of screening, at the time of initial tumor assessment, and/or at the time of study completion/premature termination of follow-up. In one embodiment, these samples are used to assess oncogene changes at baseline and to assess the likelihood of new changes occurring after treatment with inavelisib and HER2 targeted therapies.
Examples
Example 1 inhibition of proliferation of breast cancer cells
In the cell proliferation assay, we studied the effect of inanolisib and apilimbus (both PI3kα -specific inhibitors) on breast cancer cell line viability with different HER2 expression and PIK3CA mutant status. Unexpectedly, we observed a significant difference in sensitivity between inavillisib and apicalist in HER2 expanded cell line (about 20-fold difference between mean IC50 values) and HER2 negative cell line (about 6-fold difference between the two inhibitors). Regardless of HER2 status, both inhibitors did not differentiate in the wild-type (WT) cell line (fig. 1).
The cell line was obtained from ATCC. All cell lines underwent short tandem repeat analysis, SNP fingerprinting, and validation of mycoplasma testing (Yu M. Et al, "A resource for cell line authentication, annotation and quality control" Nature 520,307-311 (2015)). Cell lines were grown under standard tissue culture conditions in RPMI medium containing 10% fetal bovine serum (Gibco, 10082-147), 100U/mL penicillin-streptomycin (Gibco, 15140-122), 2mmol/L L-glutamine (Gibco, 15030-081). Cells are treated with the compound for a specified period of time.
Vitality measurement: cells (1000-2000 cells/well) were seeded in 384 well plates for 16 hours. On the next day, nine consecutive 1:3 dilutions of compound were prepared in DMSO in 96-well plates. The compounds were then further diluted into growth medium using a Rapidplate robot (Zymark corp., hopkinton, MA). The diluted compound was then added to the wells of 384 well cell plates in four replicates and at 37 ℃ and 5% CO 2 And (5) incubating. After 4 days, cellTiter-(Promega) the relative number of living cells was measured by luminescence and read on Wallac Multilabel Reader (Perkinelmer, foster City). IC (integrated circuit) 50 Calculations were performed using Prism 6.0 software (GraphPad, san Diego). Growth Rate (GR) calculations and charts were performed using R-script based on Hafner, M.et al, "Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs" Nat. Methods 13,521-527 (2016).
EXAMPLE 2 inhibition of tumor growth in vivo
The in vivo efficacy of the test compounds and combinations was tested in a KPL-4 (HER 2 positive, PIK3CA H1047R) breast cancer xenograft model. HCC1954 tumor cells (5 x 106) were inoculated into 2/3 mammary fat pads of female NCR nude mice (Taconic norm, hudson NY). Tumor volumes were measured using Ultra Cal-IV calipers (model 54-10-111;Fred V.Fowler Co; newton, mass.). The following formula was used to calculate the swelling in Excel version 11.2Tumor volume: tumor volume (mm) 3 ) = (length x width 2 ) x 0.5. Mice used for efficacy studies were divided into 8-10 mice/group with average tumor volumes of 200 to 250mm at the beginning of dosing 3 . The tumor volumes repeatedly measured over time in the same animal were analyzed using a Linear Mixed Effect (LME) modeling method (Pinheiro et al, "Linear and Nonlinear Mixed Effects Models" R package version 3;1-131 (2017)). Using a cubic regression spline to fit the log of each dose level 2 Nonlinear curve of tumor volume over time. These non-linear curves are then correlated to the dose within the hybrid model. The percent tumor growth inhibition (% TGI) as vehicle control was calculated as the percentage of area under the fitted curve (AUC) of each dose group per day relative to vehicle group using the following formula: % tgi=100 x (1-AUC dose/AUC vehicle).
Inanolisib was formulated in a methylcellulose Tween (MCT) vehicle consisting of 0.5% (w/v) methylcellulose, 0.2% (w/v) polysorbate 80 (Tween-80), and a dose of 25mg/kg was orally administered daily by gavage for 28 days. Trastuzumab and pertuzumab were formulated in HB #08 vehicle consisting of HisAc 20mM, sucr 240mM and TW-20.02% (pH 5.5), and 3mg/kg doses of trastuzumab and 2.5mg/kg pertuzumab were administered weekly by intraperitoneal (i.p.) injection for 4 weeks, respectively.
Mice were dosed in four groups (9 mice per group): group 1, vehicle (MCT); group 2, trastuzumab+pertuzumab; group 3, inavelisib; and group 4, trastuzumab+pertuzumab+inavelisib. Tumor size and mouse body weight were recorded twice weekly and tumor volume was exceeded 2000mm 3 Or mice that lost 20% of the initial body weight were immediately euthanized.
The results are shown in the following table.
In the HER2 positive mutant p110αkpl-4 xenograft model, the HER2 inhibitor combination (trastuzumab and pertuzumab) with inanolisib showed a better response than trastuzumab+pertuzumab alone or inanolisib alone.
Example 3 clinical study of inavelisib/trastuzumab/pertuzumab combinations
An open label, phase I/Ib study is ongoing with daily oral inanolisib in combination with HER 2-targeted therapy.
Approximately 20 patients may participate in dose-queue expansion (phase II, group G) to assess the safety, tolerability, pharmacokinetics and primary anti-tumor activity of inavilisib in combination with trastuzumab and pertuzumab in locally advanced or metastatic PIK3CA mutant her2+ breast cancer patients.
inanolisib (GDC-0077) is provided as a 3mg or 9mg gauge tablet. TrastuzumabProvided as a lyophilized formulation at a nominal level of 440mg per vial. Pertuzumab is provided as a disposable formulation containing 30mg/mL pertuzumab.
Study design
Locally advanced or metastatic PIK3CA mutant her2+ breast cancer patients were treated with GDC-0077 (equal to or lower than the GDC-0077MTD or MAD determined in phase I group a) in combination with trastuzumab and pertuzumab to obtain additional safety, tolerability and PK data, as well as preliminary evidence of clinical activity. In addition to treatment with GDC-0077, trastuzumab and pertuzumab, patients with hr+ disease can be treated with letrozole or fulvestrant at the discretion of the researcher and according to local guidelines.
Inavolisib was administered in combination with trastuzumab and pertuzumab (G+H+P) over a 21-day period. Patients received 3, 6 or 9mg doses of inanolisib on days 1 to 21. Patients received trastuzumab by IV infusion on day 1 of each 21-day cycle, with a loading dose of 8mg/kg for cycle 1 and a dose of 6mg/kg for the subsequent cycle; and at day 1 of each 21-day cycle pertuzumab was received by IV infusion, with a loading dose of 840mg for cycle 1 and a dose of 420mg for the subsequent cycle until disease progression or unacceptable toxicity occurred. Patients receiving trastuzumab and pertuzumab within 6 weeks of starting study treatment did not need to receive loading doses of trastuzumab and pertuzumab on day 1 of cycle 1, and received 6mg/kg trastuzumab and 420mg pertuzumab on day 1 of cycle 1 and subsequent cycles.
The baseline body weight was used to calculate the required dose of trastuzumab. If the patient's body weight changes by > 10% from baseline, the dose of trastuzumab will be recalculated. If the dose is recalculated because the body weight changes by ≡10% from the baseline, this body weight will be used as a new baseline to calculate trastuzumab doses in subsequent cycles.
Administration may be delayed to assess or treat adverse events. If the patient missed one cycle of doses of trastuzumab or pertuzumab (i.e., two sequential administrations are separated by ≡6 weeks), a reloaded dose of 8mg/kg trastuzumab or 840mg pertuzumab is administered. Subsequent doses of 420mg of pertuzumab were then administered every 21 days with 6mg/kg of trastuzumab.
On the study visit, GDC-0077 was administered first, followed by pertuzumab, and then trastuzumab. Pertuzumab and trastuzumab are administered by trained staff to monitor and respond to medical emergencies in emergency-equipped environments. Initial IV infusion of pertuzumab was administered over 60 (±10) minutes followed by a 60 minute observation period. If the initial infusion is well tolerated, the subsequent infusion may be administered over 30 (+ -10) minutes followed by a 30 minute observation period. The observation period should be completed prior to the subsequent infusion of trastuzumab. The initial IV infusion of trastuzumab was administered over 90 (±10) minutes, followed by a 30 minute observation period. If the initial infusion is well tolerated, the subsequent infusion may be administered over 30 (+ -10) minutes followed by a 30 minute observation period. At the discretion of the researcher, or if applicable, the patient may be observed for longer periods of time, as required locally. Prior to infusion of pertuzumab and trastuzumab, a precursor drug may be administered with an antipyretic, antihistamine or corticosteroid.
Inclusion criteria:
● Female participants with locally advanced or metastatic PIK3CA mutant her2+ breast cancer;
● The left ventricular ejection fraction is 50% or higher.
Key qualification criteria: PIK3CA mutations in tumor tissue or ctDNA; HER2+ is defined as HER2 IHC score 3+, HER2 IHC score 2+ with a dual probe ISH HER2/CEP17 ratio of > 2.0 (preferred), or fluorescence, chromogenic or silver ISH tests indicate the presence of HER2 gene amplification, or HER2+ according to local clinical guidelines; fasting blood glucose is less than or equal to 140mg/dL, hbA1c is less than 7%; and Left Ventricular Ejection Fraction (LVEF) > 50% determined by Echocardiography (ECHO) (preferred) or multiple gated acquisition (MUGA) scans. Patients who have received prior HER 2-targeted therapies (including trastuzumab and/or pertuzumab) have had to deactivate prior trastuzumab and/or pertuzumab due to toxicity assessed as associated with one or both agents.
Hr+ breast cancer patients may be treated with endocrine therapy (i.e., letrozole or fulvestrant) and, at the discretion of the investigator, pre-menopausal or perimenopausal patients may also be treated with LHRH agonist therapy.
Exclusion criteria:
● Current uncontrolled hypertension (systolic >150mmHg and/or diastolic >100 mmHg) or unstable angina;
● The New York Heart Association (NYHA) ranks the history of Congestive Heart Failure (CHF) of grade II or higher, or of severe cardiac arrhythmias requiring treatment (excluding atrial tremor or paroxysmal supraventricular tachycardia);
● Study the history of myocardial infarction within 6 months prior to initiation of treatment;
● Ejection fraction decreased with previous trastuzumab treatment (history of LVEF decreasing below 40% during or after previous treatment with trastuzumab);
● Exposure to cumulative doses of doxorubicin (or equivalent anthracycline exposure)>360mg/m 2 A history of body surface area or equivalent dosages thereof;
● Symptomatic active lung disease, including pneumonia or interstitial lung disease;
● The prior significant toxicity history associated with trastuzumab and/or pertuzumab that required discontinuation of treatment.
Antitumor activity was assessed at screening and every 8 weeks by RECIST v 1.1. Clinical Benefit Rate (CBR) is defined as complete or partial response, or disease stabilization lasting ≡24 weeks.
Pharmacodynamic activity was assessed by Immunohistochemistry (IHC) in tumor tissue biopsies as well as in ctDNA samples (by Foundation ACT) before and at the time of the study (2 weeks after daily inanolisib treatment).
Important safety risks of trastuzumab and pertuzumab, such as embryo-fetal toxicity, allergic/anaphylactic and infusion-related reactions, LVEF reduction, diarrhea, rash/skin reactions, stomatitis/oral mucositis and pulmonary toxicity, are managed according to the investigator manual for trastuzumab and pertuzumab.
Example 4 clinical study of inavelisib and paclitaxel with or without combination of targeted therapies
A phase Ib, open label, dose escalation and dose escalation study is ongoing that evaluates the safety, tolerability, pharmacokinetics and primary anti-tumor activity of inanolisib and paclitaxel in combination with or without targeted therapies in patients with locally advanced or metastatic solid tumors.
The objective was to evaluate the safety, pharmacokinetics, pharmacodynamic (PD) effects and preliminary anti-tumor activity of inavilisib in combination with paclitaxel in locally advanced or metastatic solid tumor patients, and of inavilisib in combination with paclitaxel, trastuzumab and pertuzumab in locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer patients.
Intervention measures
The study was open label design, and the treatment included: inanolisib-6 mg/9mg orally per day, possibly on a 6/1 or 5/2 intermittent weekly schedule, until disease progression or unacceptable toxicity occurs. Paclitaxel-80 mg/m2 dose is infused intravenously weekly until disease progression or unacceptable toxicity occurs. Trastuzumab-was infused intravenously every 3 weeks. The loading dose for cycle 1 was 8mg/kg and the loading dose for the subsequent cycle was 6mg/kg until disease progression or unacceptable toxicity occurred. Pertuzumab-was infused intravenously once every 3 weeks. The loading dose for the first cycle was 840mg and the loading dose for the subsequent cycle was 420mg until disease progression or unacceptable toxicity occurred.
Up to 120 patients from different hospitals around the world will participate in this study. The study was divided into two parts. Part 1, group a, is the dose escalation (dose finding) part of the study. Part 2 includes groups a and B, which will be the treatment extension after the completion of dose discovery. The participants were assigned to different treatment groups according to the inclusion and exclusion criteria of parts 1 and 2:
part 1, group a: inanolisib will be tested in up to 24 patients with locally advanced or metastatic cancer in combination with paclitaxel at different doses and schedules.
Part 2, group a: the inanolisib doses and schedule identified as safe in part 1 group a will be tested in combination with paclitaxel in approximately 76 locally advanced or metastatic PIK3CA mutated (genetically altered) cancer patients.
Part 2, group B: the inanolisib doses and schedule determined to be safe in group 1, part a, will be tested in approximately 20 locally advanced or metastatic PIK3CA mutated (genetically altered) HER2 positive breast cancer patients in combination with paclitaxel, trastuzumab and pertuzumab.
The patient will receive the following evaluations and measurements:
1. vital signs-body temperature, pulse rate, blood pressure, respiratory rate and blood oxygen content;
2. Comprehensive or limited physical examination;
3. physical stamina assessment (eastern tumor cooperative group daily function);
4. electrocardiography (ECG);
5. urine samples for standard laboratory tests;
6. blood samples for standard laboratory tests and to measure pharmacokinetics;
7. tumor tissue biopsies;
8. tumor assessment: visceral organ and bone scanning, may include: computed Tomography (CT) scanning, magnetic Resonance Imaging (MRI) scanning, and bone scanning;
9. ophthalmic examination.
Measurement of primary results
Security objective:
1. incidence and nature of dose limiting toxicity (part 1 only) measured using adverse events (graded by NCI CTCAE v 5.0) recorded over the first 28 days (cycle 1) of study treatment;
2. incidence, type and severity of adverse events (including serious adverse events graded by NCI CTCAE v 5.0) recorded throughout the study;
3. target vital signs measured weekly/monthly at baseline and during study treatment using standard techniques assessed by on-site staff;
4. targeted clinical laboratory test results, including ECG, measured at baseline and collected weekly/periodically, were collected and recorded by on-site staff at baseline and each period during study treatment using standard hospital laboratory analysis.
Secondary outcome measure
Pharmacokinetic targets (minor targets):
plasma concentrations of inavilisib administered in combination with paclitaxel (group a) or with paclitaxel, trastuzumab and pertuzumab (group B) were measured by the bioanalytics laboratory at cycles 1-3.
The following PK parameters will be determined as appropriate:
1. area under concentration-time curve (AUC)
2. Maximum plasma concentration (Cmax)
3. Minimum plasma concentration (Cmin)
4. Additional plasma PK parameters if necessary
Activity/efficacy targets (secondary targets):
the antitumor activity was initially evaluated as follows:
inanolisib in combination with paclitaxel in patients with localized advanced or metastatic PIK3CA mutated solid tumors (part 2 group a);
inavelisib administered in combination with paclitaxel, trastuzumab and pertuzumab in locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer patients (# part 2 group B).
The corresponding endpoints are as follows:
1. objective Remission Rate (ORR) is defined as two consecutive Complete (CR) or Partial (PR) recoveries ≡4 weeks apart, as determined by the investigator using RECIST v1.1 every 8 (×) or 9 (#) weeks during study treatment;
2. optimal total remission (BOR) is defined as the proportion of CR or PR patients, as determined by the investigator using RECIST v1.1 every 8 (×) or 9 (#) weeks during study treatment;
3. Duration of remission (DOR) is defined as the time from the first appearance of recorded objective remission to disease progression or death (based on the first occurrence) as determined by the investigator using RECIST v1.1 every 8 (×) or 9 (#) weeks during study treatment;
4. clinical Benefit Rate (CBR) is defined as the percentage of patients with non-complete remission/non-progressive disease;
5. progression Free Survival (PFS) is defined as the time from the first study treatment (day 1) to the first occurrence of disease progression or death (based on the preexisting event) as determined by the study staff using RECIST v1.1 every 8 (×) or 9 (#) weeks during the study treatment.
Participant enrollment criteria
1. Signing an informed consent form;
2. aged 18 years and older;
3. an evaluable or measurable disease according to RECIST v 1.1;
4. eastern tumor cooperative group (ECOG) physical status is 0 or 1;
5. life expectancy 12 weeks;
6. sufficient hematology and organ function was present within 14 days prior to initiation of study treatment, defined by:
6.1. absolute neutrophil count 1500/l,
6.2. Hemoglobin (hemoglobin) 9g/dl of the total weight of the product,
6.3. the platelet count was 100,000/l,
6.4. fasting blood glucose 126mg/dL or 7mmol/l and glycosylated hemoglobin (HbA 1C) 5.7%,
6.5. total bilirubin 1.5 upper normal limit (ULN),
6.6. serum albumin 2.5g/dl or 25g/l,
ast and ALT 2.5ULN, with the following exceptions: patients with liver metastasis records may have AST and ALT 5.0ULN,
6.8. serum creatinine 1.5ULN or creatinine clearance 50ml/min based on the Cockcroft-Gault glomerular filtration rate estimate;
7. consent to provide fresh (preferred) or archived tumor tissue samples;
8. for women with fertility: agreeing to maintain abstinence (avoid sexuality) or using a high-efficiency form of contraception with a failure rate of 1% per year in combination with male condoms and spermicides (male partner) unless male sterilization has been confirmed;
9. for men: either a disagreement (avoiding the sexual intercourse) or a high-efficiency contraceptive measure was agreed, and no sperm donation was agreed.
For specific inclusion criteria in recruiting patients to part 1 group a: histologically recorded, locally advanced, recurrent or metastatic, incurable solid tumor malignancy, has progressed after available standard systemic therapy; or to which standard therapies have proven ineffective or intolerable; or for which clinical trials of the agent under study are accepted standards of care. If other SOC therapies are available, they will be discussed and documented with the patient prior to informed consent.
Specific inclusion criteria for recruiting patients to part 2 group a expansion cohorts:
1. a histologically recorded, locally advanced, recurrent or metastatic, incurable solid tumor malignancy having a known PIK3CA mutation, which has progressed after at least one available standard systemic treatment in a metastatic environment;
2. confirmation of biomarker eligibility: the effective results from the blood center test or the blood or tumor tissue local test required to enroll patients in part 2 group a expansion cohort record the tumor status of PIK3CA mutations.
For specific inclusion criteria in recruiting patients to part 2 group B:
1. locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer patients with histologically recorded;
2. the patient may behave as: a new metastatic HER2 positive disease not receiving any systemic HER2 positive anti-cancer treatment, a recurrent locally advanced or metastatic disease following a previous HER2 positive targeting treatment for early breast cancer, wherein the diagnosis is made based on a biopsy of the locally recurrent or metastatic disease, and the patient progresses after receiving (new) adjuvant HER2 positive targeting therapy with a 6 month no treatment interval;
3. HER2 positive and HR positive or HR negative breast cancers recorded according to ASCO/CAP guidelines based on HER2 positive local assessment;
4. confirmation of biomarker eligibility: effective results from the blood center test required to enroll patients in part 2 group B, which record the tumor status of PIK3CA mutations;
5. at screening, left Ventricular Ejection Fraction (LVEF) 50% determined by Echocardiography (ECHO) (preferred) or multiple gated acquisition (MUGA) scans;
6. for women with fertility: agreeing to maintain abstinence (avoiding sexuality) or to use a high-efficiency form of contraceptive method with a failure rate of 1% per year during treatment and following periods, and agreeing not to donate eggs: at least 60 days after the last dose of inavelisib, at least 6 months after the last dose of paclitaxel, at least 7 months after the last dose of pertuzumab and the last dose of trastuzumab.
Participant exclusion criteria
1. Metaplasia breast cancer;
2. any history of leptomeningeal disease;
3. type 2 diabetes requiring sustained systemic treatment upon entry into the study; or any history of type 1 diabetes;
4. inability or unwilling to swallow pills;
5. malabsorption syndrome or other conditions that interfere with intestinal absorption;
6. Known and untreated or active CNS metastasis (progression, or need anticonvulsants or corticosteroids to control symptoms);
7. uncontrolled pleural effusion or ascites requiring repeated drainage twice a month or more;
8. researchers believe that any active infection affects patient safety; alternatively, a severe infection requiring IV infusion of antibiotics within 7 days prior to day 1 of cycle 1;
9. any concurrent ocular or intraocular disorder (e.g., cataract or diabetic retinopathy) appears to the researcher or research ophthalmologist to require pharmaceutical or surgical intervention during the research to prevent or treat vision loss that may be caused by the disorder;
10. active inflammation (e.g., uveitis or vitreoitis) or an infectious condition (e.g., conjunctivitis, keratitis, scleritis, or endophthalmitis) of either eye or spontaneous or autoimmune related uveitis history of either eye;
11. patients requiring daily oxygen supplementation;
12. active inflammation (e.g., crohn's disease or ulcerative colitis) or any active intestinal inflammation (including diverticulitis) history or current state;
13. patients currently receiving immunosuppressants (e.g., sulfasalazine) are considered to have active disease; therefore, they are not acceptable;
14. Symptomatic hypercalcemia requiring continued treatment with bisphosphonates or diels-semab (denosumab);
15. a clinically significant history of liver disease, including severe liver injury (Child-Pugh B/C grade), viral hepatitis or other hepatitis, current alcoholism or cirrhosis;
16. known HIV infection;
17. any other disease, active or uncontrolled pulmonary dysfunction, metabolic dysfunction, physical examination findings or clinical laboratory examination findings reasonably suspects a disease or condition that prohibits the use of study drugs, may affect the interpretation of the results or put the patient at high risk of developing therapeutic complications;
18. significant traumatic injury or significant surgery occurred within 4 weeks prior to initiation of study treatment;
19. radiation therapy (other than palliative radiation therapy for bone metastases) was received as a cancer therapy within 4 weeks prior to initiation of study treatment;
20. palliative irradiation for bone metastases was performed within 2 weeks prior to initiation of study treatment;
21. unresolved toxicity from previous therapies, except for the following: level 1 alopecia and peripheral neuropathy;
22. failing to follow study and follow-up procedures;
23. other malignancy history within 5 years prior to screening, except for malignancy that has negligible risk of metastasis or death and/or is treated to have an expected cure outcome (e.g., fully treated carcinoma in situ of the cervix, non-melanoma skin cancer, localized prostate cancer, ductal carcinoma in situ, or stage I uterine cancer);
24. Active ventricular arrhythmias or congestive heart failure or history or current status of symptomatic coronary heart disease requiring medication;
25. clinically significant electrolyte abnormalities (e.g., hypokalemia, hypomagnesemia, hypocalcemia);
26. the congenital long QT syndrome group or QT interval, as evidenced by at least two electrocardiograms spaced 30 minutes apart, is corrected for 470ms with the friedrisiella formula (QTcF), or for family history of sudden death or long QT syndrome of unknown cause;
27. currently receiving the administration of known prolongation QT interval;
28. allergic or anaphylactic reactions to the inanolisib formulation ingredients and paclitaxel;
29. pregnancy, lactation or the intention of pregnancy or child birth during the study period;
30. women with fertility, including those who have had oviduct ligators, must have a negative serum pregnancy test result within 14 days prior to initiation of study treatment.
For specific exclusion criteria in recruiting patients to part 1 group a:
1. prior significant toxicity history associated with PI3K, AKT or mTOR inhibitors, requires cessation of treatment. The patient may have received prior treatment with PI3K, AKT or mTOR inhibitors;
2. prior significant toxicity history associated with paclitaxel treatment, the treatment needs to be discontinued. The patient may have received prior treatment with paclitaxel;
3. Treatment with chemotherapy, immunotherapy or biologic therapy as an anticancer therapy was received within 21 days prior to the initiation of study treatment, except for the following: regulatory agency approved kinase inhibitors may be used up to 2 weeks prior to initiation of study treatment, provided that any drug-related toxicity has resolved to up to grade 1 and prior to approval by the medical inspector. Treatment with the agents in the study was performed within 3 weeks or five half-lives before the study treatment began, whichever was shorter. Shorter clearance periods may be allowed if the patient has adequately recovered from any clinically relevant toxicity and obtained prior approval by a medical supervisor.
4. Previous anti-cancer therapies meeting the following criteria: high dose chemotherapy requiring stem cell support; the 25% bone marrow containing area was irradiated.
For specific exclusion criteria in recruiting patients to part 2 group a:
1. previous significant toxicity history associated with PI3K, AKT or mTOR inhibitors, requiring cessation of treatment;
2. previous treatments with any PI 3K-specific inhibitor;
3. prior significant toxicity history associated with paclitaxel treatment, the treatment needs to be discontinued. The patient may have received prior treatment with paclitaxel;
4. Treatment with chemotherapy, immunotherapy or biologic therapy as an anticancer therapy was received within 21 days prior to the initiation of study treatment, except for the following: regulatory agency approved kinase inhibitors may be used up to 2 weeks prior to initiation of study treatment, provided that any drug-related toxicity has resolved to up to grade 1 and prior to approval by a medical supervisor; treatment with the agent in the study was performed within 3 weeks or five half-lives before the study treatment began, whichever was shorter;
5. previous anticancer therapies meeting the following criteria: high dose chemotherapy requiring stem cell support; 25% of the bone marrow area was irradiated.
Specific exclusion criteria for recruiting patients to part 2 group B:
1. previous treatments with any PI3K, AKT or mTOR inhibitor or with any agent that acts by a mechanism that inhibits the PI3K/AKT/mTOR pathway;
2. prior to initiation of study treatment, any prior systemic anti-cancer treatment against locally advanced or metastatic HER2 positive breast cancer;
3. currently uncontrolled hypertension (systolic greater than 150mmHg or diastolic greater than 100 mmHg) or unstable angina;
4. the New York Heart Association (NYHA) ranks the history of Congestive Heart Failure (CHF) of grade II or higher, or of severe cardiac arrhythmias requiring treatment (excluding atrial tremor or paroxysmal supraventricular tachycardia);
5. Study the history of myocardial infarction within 6 months prior to initiation of treatment;
6. during or after prior treatment with trastuzumab, LVEF drops to a history of less than 40%;
7. exposure to cumulative doses of doxorubicin (or equivalentAnthracycline exposure) 360mg/m 2 A history of body surface area or equivalent dosages thereof;
8. symptomatic active lung disease, including pneumonia or interstitial lung disease;
9. the past history of significant toxicity associated with paclitaxel, trastuzumab or pertuzumab, requires discontinuation of treatment.
Many modifications and other embodiments of the inventions set forth herein will come to mind to one skilled in the art to which these inventions pertain having the benefit of the teachings presented in the foregoing descriptions and the associated drawings. Therefore, it is to be understood that the inventions are not to be limited to the specific embodiments disclosed and that modifications and other embodiments are intended to be included within the scope of the appended claims. Although specific terms are employed herein, they are used in a generic and descriptive sense only and not for purposes of limitation.

Claims (17)

1. A method of treating HER2 positive breast cancer in a patient having locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer, the method comprising administering to the patient a combination therapy comprising:
(i)inavolisib;
(ii) Trastuzumab;
(iii) Pertuzumab; and optionally
(iv) Paclitaxel is used as the active ingredient of the medicine,
wherein the combination therapy is administered over a 21 day period.
2. A method of treating HER2 positive breast cancer in a patient having locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. QDs were administered inavilisib on days 1 to 21 of the first 21 day cycle;
b. trastuzumab was administered on day 1 of the first 21-day cycle; and
c. pertuzumab was administered on day 1 of the first 21 day cycle.
3. A method of treating HER2 positive breast cancer in a patient having locally advanced or metastatic PIK3CA mutated HER2 positive breast cancer, the method comprising administering to the patient a combination therapy comprising a dosing regimen comprising:
a. paclitaxel was administered on days 1, 8 and 15 of the first 21-day cycle;
b. QDs were administered inavilisib on days 2 to 21 of the first 21 day cycle;
c. trastuzumab was administered on day 2 of the first 21-day cycle;
d. pertuzumab was administered on day 2 of the first 21-day cycle;
And the dosing regimen further comprises up to five additional 21-day cycles comprising:
e. paclitaxel was administered on days 1, 8 and 15 of each additional 21-day cycle;
f. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
g. trastuzumab was administered on day 1 of each additional 21-day cycle; and
h. pertuzumab was administered on day 1 of each additional 21-day cycle.
4. A method according to any one of claims 1 to 3, further comprising one or more additional 21-day cycles comprising:
a. inavelisib was administered on days 1 to 21 of each additional 21-day cycle;
b. trastuzumab was administered on day 1 of each additional 21-day cycle; and
c. pertuzumab was administered on day 1 of each additional 21-day cycle.
5. The method of any one of claims 1 to 4, wherein inanolisib is administered in an amount of 9 mg.
6. The method of claim 5, wherein inanolisib is administered in an amount of 9mg in an oral tablet.
7. The method of any one of claims 1 to 6, wherein trastuzumab is administered by Intravenous (IV) infusion at a loading dose of 8mg/kg for the first 21-day period and at a dose of 6mg/kg for each additional 21-day period.
8. The method of any one of claims 1-7, wherein pertuzumab is administered by IV infusion at a loading dose of 840mg for the first 21-day period and at a dose of 420mg for each additional 21-day period.
9. The method of any one of claims 1 to 8, wherein paclitaxel is at 80mg/m for the first 21-day period and at most five additional 21-day periods 2 Is administered by IV infusion.
10. The method of any one of claims 1 to 9, wherein the patient has a Left Ventricular Ejection Fraction (LVEF) of 50% or greater.
11. The method of any one of claims 1 to 10, wherein the patient has HER2 positive breast cancer with hormone receptor positive (hr+) locally advanced or metastatic PIK3CA mutations.
12. The method of claim 11, wherein the method further comprises administering fulvestrant to the patient.
13. The method of claim 12, wherein fulvestrant is administered by Intramuscular (IM) infusion at a dose of 500mg about once every four weeks.
14. The method of claim 11, wherein the method further comprises administering letrozole to the patient.
15. The method of claim 14, wherein letrozole is administered daily in an oral tablet at a dose of 2.5 mg.
16. A method of inhibiting tumor growth or producing/increasing tumor regression in a patient suffering from HER2 positive breast cancer with locally advanced or metastatic PIK3CA mutation, the method comprising administering a combination therapy to the patient according to the method of any one of claims 1 to 15.
17. A method of preventing or delaying development of resistance of breast cancer to HER 2-targeted therapy, the method comprising administering a combination therapy comprising inanolisib, trastuzumab and pertuzumab according to the method of any one of claims 1-16.
CN202180083440.6A 2020-12-11 2021-12-07 Combination therapy for the treatment of HER2 cancer Pending CN116583540A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US63/124,495 2020-12-11
US63/161,153 2021-03-15
US202163209302P 2021-06-10 2021-06-10
US63/209,302 2021-06-10
PCT/US2021/062101 WO2022125483A1 (en) 2020-12-11 2021-12-07 Combination therapies for treatment of her2 cancer

Publications (1)

Publication Number Publication Date
CN116583540A true CN116583540A (en) 2023-08-11

Family

ID=87536484

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180083440.6A Pending CN116583540A (en) 2020-12-11 2021-12-07 Combination therapy for the treatment of HER2 cancer

Country Status (1)

Country Link
CN (1) CN116583540A (en)

Similar Documents

Publication Publication Date Title
JP6388859B2 (en) Combination and use of anti-HER2 antibody-drug conjugate and chemotherapeutic agent
US12048698B2 (en) Treatment of HER2 positive cancers
US20230310455A1 (en) Combination therapies for treatment of her2 cancer
TW202027736A (en) Methods of treating cancer with pi3k alpha inhibitors and metformin
TW202011966A (en) Methods of treating cancer with pi3k inhibitor, gdc-0077
CN114945369A (en) Methods of treating HER2 positive breast cancer with a combination of Tucotinib and an anti-HER 2 antibody-drug conjugate
US20230088701A1 (en) Combination therapies for treatment of breast cancer
JP2022553041A (en) Methods of treating HER2-positive breast cancer with tucatinib in combination with capecitabine and trastuzumab
CN116583540A (en) Combination therapy for the treatment of HER2 cancer
US20190211102A1 (en) Methods and combination therapy to treat cancer
WO2023230429A1 (en) Methods of treating colorectal cancer with tucatinib in combination with an anti-her2 antibody
EA044960B1 (en) TREATMENT OF HER2-POSITIVE CANCER
TW202430189A (en) Methods of treating cancer with pi3k inhibitor, gdc-0077

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40092971

Country of ref document: HK