Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sarat Chandarlapaty is active.

Publication


Featured researches published by Sarat Chandarlapaty.


Science Translational Medicine | 2015

PI3K inhibition results in enhanced estrogen receptor function and dependence in hormone receptor–positive breast cancer

Ana Bosch; Zhiqiang Li; Anna Bergamaschi; Haley Ellis; Eneda Toska; Aleix Prat; Jessica J. Tao; Daniel E. Spratt; Nerissa Viola-Villegas; Pau Castel; Gerard Minuesa; Natasha Morse; Jordi Rodon; Yasir H. Ibrahim; Javier Cortes; Jose Perez-Garcia; Patricia Galván; Judit Grueso; Marta Guzman; John A. Katzenellenbogen; Michaelz Kharas; Jason S. Lewis; Maura N. Dickler; Violeta Serra; Neal Rosen; Sarat Chandarlapaty; Maurizio Scaltriti; J. Baselga

Inhibition of the PI3K/AKT pathway results in induction of ER-dependent transcriptional activity and susceptibility to anti-estrogen therapy in ER-positive breast cancer. PIKing the correct therapeutic combination Mutations in a gene called PIK3CA are very common in estrogen receptor–positive breast cancers, and drugs that inhibit PI3K, the protein product of this gene, are already in clinical development. Unfortunately, these drugs are not always effective, and this study by Bosch et al. demonstrates a reason for this problem and a practical way to overcome it. By studying both mouse models and human patients’ tumors, the authors discovered that inhibition of PI3K often stimulates the activity of the estrogen receptor, which then drives tumor growth. By combining PI3K inhibitors with clinically available drugs that inhibit the estrogen receptor, the authors were able to overcome treatment resistance and effectively induce tumor regression in mouse models. Activating mutations of PIK3CA are the most frequent genomic alterations in estrogen receptor (ER)–positive breast tumors, and selective phosphatidylinositol 3-kinase α (PI3Kα) inhibitors are in clinical development. The activity of these agents, however, is not homogeneous, and only a fraction of patients bearing PIK3CA-mutant ER-positive tumors benefit from single-agent administration. Searching for mechanisms of resistance, we observed that suppression of PI3K signaling results in induction of ER-dependent transcriptional activity, as demonstrated by changes in expression of genes containing ER-binding sites and increased occupancy by the ER of promoter regions of up-regulated genes. Furthermore, expression of ESR1 mRNA and ER protein were also increased upon PI3K inhibition. These changes in gene expression were confirmed in vivo in xenografts and patient-derived models and in tumors from patients undergoing treatment with the PI3Kα inhibitor BYL719. The observed effects on transcription were enhanced by the addition of estradiol and suppressed by the anti-ER therapies fulvestrant and tamoxifen. Fulvestrant markedly sensitized ER-positive tumors to PI3Kα inhibition, resulting in major tumor regressions in vivo. We propose that increased ER transcriptional activity may be a reactive mechanism that limits the activity of PI3K inhibitors and that combined PI3K and ER inhibition is a rational approach to target these tumors.


Cancer Cell | 2015

Feedback Suppression of PI3Kα Signaling in PTEN-Mutated Tumors Is Relieved by Selective Inhibition of PI3Kβ

Sarit Schwartz; John Wongvipat; Cath Trigwell; Urs Hancox; Brett S. Carver; Vanessa Rodrik-Outmezguine; Marie Will; Paige Yellen; Elisa de Stanchina; José Baselga; Howard I. Scher; Simon T. Barry; Charles L. Sawyers; Sarat Chandarlapaty; Neal Rosen

In PTEN-mutated tumors, we show that PI3Kα activity is suppressed and PI3K signaling is driven by PI3Kβ. A selective inhibitor of PI3Kβ inhibits the Akt/mTOR pathway in these tumors but not in those driven by receptor tyrosine kinases. However, inhibition of PI3Kβ only transiently inhibits Akt/mTOR signaling because it relieves feedback inhibition of IGF1R and other receptors and thus causes activation of PI3Kα and a rebound in downstream signaling. This rebound is suppressed and tumor growth inhibition enhanced with combined inhibition of PI3Kα and PI3Kβ. In PTEN-deficient models of prostate cancer, this effective inhibition of PI3K causes marked activation of androgen receptor activity. Combined inhibition of both PI3K isoforms and androgen receptor results in major tumor regressions.


Clinical Breast Cancer | 2014

A Phase II Open-Label Study of Ganetespib, a Novel Heat Shock Protein 90 Inhibitor for Patients With Metastatic Breast Cancer

Komal Jhaveri; Sarat Chandarlapaty; Diana Lake; Teresa Gilewski; Mark Robson; Shari Goldfarb; Pamela Drullinsky; Steven Sugarman; Carolyn Wasserheit-Leiblich; Julie Fasano; Mary Ellen Moynahan; Gabriella D'Andrea; Kristina Lim; Laura Reddington; Sofia Haque; Sujata Patil; Lynne Bauman; Vojo Vukovic; Iman El-Hariry; Clifford A. Hudis; Shanu Modi

BACKGROUNDnGanetespib is a small molecule, nongeldanamycin HSP90 inhibitor with potent inhibitory effects on HSP90-dependent oncoproteins of relevance to breast cancer pathogenesis. We therefore tested ganetespib in an unselected cohort of patients with MBC.nnnPATIENTS AND METHODSnPatients were treated with single agent ganetespib at 200 mg/m(2) once weekly for 3 weeks, on a 28-day cycle. Therapy was continued until disease progression. The primary end point was ORR using Reponse Evaluation Criteria in Solid Tumors version 1.1.nnnRESULTSnTwenty-two patients were enrolled with a median age of 51(range, 38-70) years and a median Eastern Cooperative Oncology Group performance status of 0 (range, 0-1). Most patients had at least 2 previous lines of chemotherapy in the metastatic setting. Most common toxicities, largely grade 1/2, were diarrhea, fatigue, nausea, and hypersensitivity reaction. The ORR in this unselected population was 9%, with all responses coming from the subset of patients with HER2-positive MBC (2/13;xa015%). One patient with TNBC had objective tumor regression in the lung metastases. The clinical benefit rate (complete responsexa0+ partial responsexa0+ stable disease > 6 months) was 9%, median progression-free survival was 7xa0weeks (95% confidence interval [CI], 7-19), and median overall survival was 46 weeks (95% CI, 27-not applicable).nnnCONCLUSIONnThe study did not meet the prespecified criteria for ORR in the first stage of the Simon 2-stage model in this heavily pretreated unselected population of MBC. However, activity was observed in trastuzumab-refractory HER2-positive and TNBC. Ganetespib was well tolerated and responses in more targeted populations harboring specific HSP90-dependent oncoproteins justifies its further study, particularly as part of rational combinations.


The Journal of Nuclear Medicine | 2016

Detection of HER2-Positive Metastases in Patients with HER2-Negative Primary Breast Cancer Using 89Zr-Trastuzumab PET/CT

Gary A. Ulaner; David M. Hyman; Dara S. Ross; Adriana D. Corben; Sarat Chandarlapaty; Shari Goldfarb; Heather L. McArthur; Joseph P. Erinjeri; Stephen B. Solomon; Hartmuth C. Kolb; Serge K. Lyashchenko; Jason S. Lewis; Jorge A. Carrasquillo

Our objective was to determine whether imaging with a human epidermal growth factor receptor 2 (HER2)–targeted PET tracer can detect HER2-positive metastases in patients with HER2-negative primary breast cancer. Methods: Patients with HER2-negative primary breast cancer and evidence of distant metastases were enrolled in an Institutional Review Board–approved prospective clinical trial. Archived pathologic samples from the patient’s primary breast cancer were retested to confirm HER2-negative disease. Patients with confirmed HER2-negative primary breast cancer underwent 89Zr-trastuzumab PET/CT to screen for 89Zr-trastuzumab metastases. Metastases avid for 89Zr-trastuzumab by PET/CT were biopsied and pathologically examined to define HER2 status. Patients with pathologically proven HER2-positive metastases subsequently received off-protocol HER2-targeted therapy to evaluate treatment response. Results: Nine patients were enrolled, all of whom had pathologic retesting that confirmed HER2-negative primary breast cancer. Five demonstrated suggestive foci on 89Zr-trastuzumab PET/CT. Of these 5 patients, 2 had biopsy-proven HER2-positive metastases and went on to benefit from HER2-targeted therapy. In the other 3 patients, biopsy showed no evidence of HER2-positive disease, and their foci on 89Zr-trastuzumab PET were considered false-positive. Conclusion: In this proof-of-concept study, we demonstrated that 89Zr-trastuzmab PET/CT detects unsuspected HER2-positive metastases in patients with HER2-negative primary breast cancer. Although these are only initial results in a small sample, they are a proof of the concept that HER2-targeted imaging can identify additional candidates for HER2-targeted therapy. More specific HER2-targeted agents will be needed for clinical use.


Oncogene | 2017

Acquired CDK6 amplification promotes breast cancer resistance to CDK4/6 inhibitors and loss of ER signaling and dependence

C Yang; Z Li; T Bhatt; M Dickler; D Giri; Maurizio Scaltriti; Josep Baselga; Neal Rosen; Sarat Chandarlapaty

Dysregulated activation of the CDK4/6 kinases is a hallmark of most mammary-derived carcinomas. ATP-competitive inhibitors against this complex have been recently advanced in the clinic and have shown significant activity, particularly against tumors driven by the estrogen receptor (ER). However, resistance to these compounds has begun to emerge often months to years after their initiation. We investigated potential mechanisms of resistance using cell line models that are highly sensitive to this class of drugs. After prolonged exposure to the selective and potent CDK4/6 inhibitor LY2835219, clones emerged and several were found to harbor amplification of the CDK6 kinase. Amplification of CDK6 resulted in a marked increase in CDK6 expression and reduced response of the CDK4/6 target, phospho-Rb (pRb), to CDK4/6 inhibitors. Knockdown of CDK6 restored drug sensitivity, while enforced overexpression of CDK6 was sufficient to mediate drug resistance. Not only did CDK6 overexpression mediate resistance to CDK4/6 inhibitors but it also led to reduced expression of the ER and progesterone receptor (PR), and diminished responsiveness to ER antagonism. The reduced ER/PR expression after CDK4/6 inhibitor resistance was additionally observed in tumor biopsy specimens from patients treated with these drugs. Alternative mechanisms of resistance to CDK4/6 inhibitors such as loss of pRb and cyclin E1 overexpression also exhibited decreased hormone responsiveness, suggesting that the clinical paradigm of sequential endocrine-based therapy may be ineffective in some settings of acquired CDK4/6 resistance.


Oncogene | 2018

KMT2C mediates the estrogen dependence of breast cancer through regulation of ERα enhancer function

Kinisha Gala; Qing Li; Amit U. Sinha; Pedram Razavi; Madeline Dorso; Francisco Sanchez-Vega; Young Rock Chung; Ronald C. Hendrickson; James J. Hsieh; Michael F. Berger; Nikolaus Schultz; Alessandro Pastore; Omar Abdel-Wahab; Sarat Chandarlapaty

Estrogen receptor alpha (ERα) is a ligand-activated nuclear receptor that directs proliferation and differentiation in selected cancer cell types including mammary-derived carcinomas. These master-regulatory functions of ERα require trans-acting elements such as the pioneer factor FOXA1 to establish a genomic landscape conducive to ERα control. Here, we identify the H3K4 methyltransferase KMT2C as necessary for hormone-driven ERα activity and breast cancer proliferation. KMT2C knockdown suppresses estrogen-dependent gene expression and causes H3K4me1 and H3K27ac loss selectively at ERα enhancers. Correspondingly, KMT2C loss impairs estrogen-driven breast cancer proliferation but has no effect on ER- breast cells. Whereas KMT2C loss disrupts estrogen-driven proliferation, it conversely promotes tumor outgrowth under hormone-depleted conditions. In accordance, KMT2C is one of the most frequently mutated genes in ER-positive breast cancer with KMT2C deletion correlating with significantly shorter progression-free survival on anti-estrogen therapy. From a therapeutic standpoint, KMT2C-depleted cells that develop hormone-independence retain their dependence on ERα, displaying ongoing sensitivity to ERα antagonists. We conclude that KMT2C is a key regulator of ERα activity whose loss uncouples breast cancer proliferation from hormone abundance.


Cancer | 2018

Feasibility, safety, and efficacy of aerobic training in pretreated patients with metastatic breast cancer: A randomized controlled trial: Exercise in Metastatic Breast Cancer

Jessica M. Scott; Neil M. Iyengar; Tormod S. Nilsen; Meghan Michalski; Samantha Thomas; James E. Herndon; John Sasso; Anthony F. Yu; Sarat Chandarlapaty; Chau T. Dang; Elizabeth A. Comen; Maura N. Dickler; Jeffrey Peppercorn; Lee W. Jones

The investigation of exercise training in metastatic breast cancer has received minimal attention. This study determined the feasibility and safety of aerobic training in metastatic breast cancer.


Journal of Vascular and Interventional Radiology | 2018

Association of PI3K Pathway Mutations with Early Positron-Emission Tomography/CT Imaging Response after Radioembolization for Breast Cancer Liver Metastases: Results of a Single-Center Retrospective Pilot Study

Amy R. Deipolyi; Christopher C. Riedl; Jacqueline Bromberg; Sarat Chandarlapaty; Christopher A. Klebanoff; C. Sofocleous; Hooman Yarmohammadi; Lynn A. Brody; F. Edward Boas; Etay Ziv

PURPOSEnTo describe imaging response and survival after radioembolization for metastatic breast cancer and to delineate genetic predictors of imaging responses and outcomes.nnnMATERIALS AND METHODSnThis retrospective study included 31 women (average age, 52 y) with liver metastasis from invasive ductal carcinoma who underwent resin and glass radioembolization (average cumulative dose, 2.0 GBq ± 1.8) between January 2011 and September 2017 after receiving ≥ 3 lines of chemotherapy. Twenty-four underwent genetic profiling with MSK-IMPACT or Sequenom; 26 had positron-emission tomography (PET)/CT imaging before and after treatment. Survival after the first radioembolization and 2-4-month PET/CT imaging response were assessed. Laboratory and imaging features were assessed to determine variables predictive of outcomes. Unpaired Student t tests and Fisher exact tests were used to compare responders and nonresponders categorized by changes in fluorodeoxyglucose avidity. Kaplan-Meier survival analysis was used to determine the impact of predictors on survival after radioembolization.nnnRESULTSnMedian survival after radioembolization was 11 months (range, 1-49 mo). Most patients (18 of 26; 69%) had complete or partial response based on changes in fluorodeoxyglucose avidity. Imaging response was associated with longer survival (Pxa0= .005). Whereas 100% of patients with PI3K pathway mutations showed an imaging response, only 45% of wild-type patients showed axa0response (Pxa0= .01). Median survival did not differ between PI3K pathway wild-type (10.9 mo) and mutant (undefined) patients (Pxa0=xa0.50).nnnCONCLUSIONSnThese preliminary data suggest that genomic profiling may predict which patients with metastatic breast cancer benefit most from radioembolization. PI3K pathway mutations are associated with improved imaging response, which is associated with longer survival.


Cancer Research | 2016

Abstract P4-14-24: Optimization of intermittent high dose lapatinib administration with or without capecitabine: A rational approach to drug dosing and scheduling using Norton-Simon modeling

A Morikawa; E De Stanchina; S Patil; Sarat Chandarlapaty; Bob T. Li; Larry Norton; Andrew D. Seidman

Background: Systemic treatment of central nervous system (CNS) metastases remains a challenge partially due to poor drug penetration. Lapatinib and capecitabine are drugs with modest efficacy in treatment of brain metastases from HER2-positive (+) breast cancer (BC) and were shown to cross the blood-tumor barrier in clinical craniotomy specimens (Lin N et al., CCR 2009, Morikawa A et al., Neuro Oncol 2015). However, intratumoral drug concentrations observed were sub-optimal and heterogeneous. Administration of shorter-duration, high dose tyrosine kinase inhibitor is proposed as a way to improve efficacy and tolerability based on Norton-Simon modeling and drug exposure in the CNS (Traina T et al., JCO 2008, Grommes C et al., Neuro Oncol 2011, Chien AJ et al., J Clin Oncol 2014) . In this study, we examined optimization of high dose lapatinib administration with or without capecitabine to inform the design of a phase I trial for BC patients with HER2+ CNS metastases. Methods: Mice bearing BT-474 BC xenograft tumors were treated with various lapatinib doses and schedules. A standard continuous daily dose (100mg/kg) was compared to various intermittent dosing schedules (at 100mg/kg, 400mg/kg, and 800mg/kg). In addition, high dose lapatinib (800mg/kg) was administered with capecitabine either concurrently or in tandem. Xenografts were treated when tumors reached 100mm3. Tumor volumes were evaluated for antitumor efficacy, and mice weights were measured for toxicity. Significance testing for between-group comparisons was conducted using a mixed effect model for repeated measures. Results: Intermittent schedules of lapatinib at 100mg/kg given as 3 days on/11 days off (3/11), 5 days on/9 days off (5/9), and 7 days on/7days off (7/7) had a similar efficacy in tumor control: percent change in tumor volume of 225% (7/7), 222% (5/9), and 223% (3/11) (NS). Therefore, the 3 days on (with 4 days off or 11 days off ) schedule was subsequently chosen to evaluate for tolerability and antitumor efficacy of higher lapatinib dose. The 3 days on/4 days off (3/4) group at 800mg/kg demonstrated the highest tumor reduction (-69%) compared to the daily continuous dosing group (-18%) (p=0.04), but a trend toward higher toxicity was observed (p=0.12). Evaluation of concurrent vs. tandem administration of capecitabine with lapatinib at 800mg/kg given in 3 days on/11 days off was conducted. The concurrent treatment was discontinued early due to high toxicity. However, tandem administration of capecitabine with high dose lapatinib was tolerable without a significant difference in weight changes (p=0.62). Conclusions: The intermittent schedule allows delivery of high dose lapatinib, which has better anti-tumor activity than standard continuous dosing. If given intermittently, high dose lapatinib is tolerable, even with capecitabine if given in tandem/sequence. Based on the result of these experiments, a phase I trial of high dose lapatinib using 3 days on/11 days off schedule in tandem with capecitabine is currently proposed for treatment of HER2-positive BC patient with CNS metastases. Citation Format: Morikawa A, De Stanchina E, Patil S, Chandarlapaty S, Li BT, Norton L, Seidman AD. Optimization of intermittent high dose lapatinib administration with or without capecitabine: A rational approach to drug dosing and scheduling using Norton-Simon modeling. [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P4-14-24.


In: AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 05 Nov 2015-09 Dec 2015; Hynes Convention Center, Boston, Massachusetts, USA. http://www.abstractsonline.com/plan/start.aspx?mkey=%7bC69F0C7B-CFF2-4010-AC6C-DEAE12E60491%7d; 2015. | 2015

AZD5363, a catalytic pan-Akt inhibitor, in Akt1 E17K mutation positive advanced solid tumors

David M. Hyman; Lillian Mary Smyth; Philippe L. Bedard; Amit M. Oza; Emma Dean; Anne C Armstrong; Joao Paulo Lima; Hideaki Bando; Peter Kabos; J. Alejandro Pérez-Fidalgo; Kathleen N. Moore; Shannon N. Westin; Benoit You; Sarat Chandarlapaty; Leila Alland; Helen Ambrose; Andrew Foxley; Justin Lindemann; Martin Pass; Paul Rugman; Shaista Salim; Gaia Schiavon; K Tamura; J. Baselga; Udai Banerji

Collaboration


Dive into the Sarat Chandarlapaty's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy R. Deipolyi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

C. Sofocleous

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher A. Klebanoff

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christopher C. Riedl

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David M. Hyman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Etay Ziv

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hooman Yarmohammadi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacqueline Bromberg

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge