1
|
Expert Consensus on the Clinical Application of PI3K/AKT/mTOR Inhibitors in the Treatment of Breast Cancer (2025 Edition). CANCER INNOVATION 2025; 4:e70008. [PMID: 40206206 PMCID: PMC11981814 DOI: 10.1002/cai2.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Background The phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB or AKT)/mammalian target of rapamycin (mTOR) signaling pathway (PAM pathway) plays a critical role in breast cancer pathogenesis and progression, and is closely linked with resistance to endocrine therapy in advanced breast cancer. Randomized clinical trials have shown that PI3K/AKT/mTOR inhibitors deliver significant clinical benefits, particularly for patients with advanced hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer. Methods In 2022, the Breast Cancer Expert Committee of the National Cancer Quality Control Center convened specialists in related fields to draft the "Expert Consensus on the Clinical Application of PI3K/AKT/mTOR Inhibitors in the Treatment of Advanced Breast Cancer." This consensus raised awareness of these inhibitors among oncologists in China and improved the precision of clinical decision-making. In recent years, growing evidence has emphasized the importance of targeting the PAM pathway, reflected in the approval of several innovative agents. This consensus is an updated 2025 edition that retains the foundational structure of the 2022 edition while incorporating notable updates. Results Updates to the consensus include the introduction of newly approved PAM pathway inhibitors, updated data from recent clinical trials, and expanded therapeutic applications. The revised guidance also offers updated recommendations for genetic testing to detect alterations in relevant pathways. The section on managing drug-related adverse events has been significantly expanded, providing detailed insights into different types of adverse events and their management. These updates aim to enhance the clinical application of PAM pathway inhibitors, promote precision medicine, and ultimately, improve survival outcomes for patients with breast cancer.
Collapse
|
2
|
Galuia M, Fedorova J, McHayleh W, Mamounas E, Ahmad S, Pavri S. Perioperative Drug Management of Systemic Therapies in Breast Cancer: A Literature Review and Treatment Recommendations. Curr Oncol 2025; 32:154. [PMID: 40136358 PMCID: PMC11941735 DOI: 10.3390/curroncol32030154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer accounts for about 30% of all new female cancers each year, and its incidence is increasing 0.6% per year. An enhanced understanding of the molecular mechanisms of carcinogenesis has led to the development of constantly evolving strategies for local and systemic therapies. Perioperative chemotherapy, immunotherapy, and endocrine therapy play pivotal roles in the overall treatment plan. Guidelines on the appropriate use of these drugs in patients undergoing extirpative breast surgery and/or breast reconstruction are lacking. Clear indications for the management of systemic therapies relative to the timing of surgery is crucial to ensure consistent treatment outcomes and to minimize complications. Our purpose is to propose evidence-based recommendations to optimize the perioperative management of systemic therapies in patients undergoing breast cancer surgery and breast reconstructive surgery. In this review, we outline the basic tenets of breast cancer therapies, provide an overview on wound-healing principles, delineate relevant pharmacodynamic concepts, summarize literature and pharmacologic data from various preclinical studies and clinical trials, and propose treatment recommendations. Synopsis: This review proposes evidence-based recommendations regarding systemic therapies management for outcome optimization in the perioperative period in breast cancer patients.
Collapse
Affiliation(s)
- Mariem Galuia
- Department of Internal Medicine, AdventHealth Hospital, Orlando, FL 32804, USA;
| | - Julia Fedorova
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| | - Wassim McHayleh
- Department of Medical Oncology, AdventHealth Hospital, Altamonte Springs, FL 32701, USA
| | - Eleftherios Mamounas
- Division of Breast Surgery, Department of Surgery, AdventHealth Hospital, Orlando, FL 32804, USA;
| | - Sarfraz Ahmad
- Gynecologic Oncology Program, AdventHealth Cancer Institute, Orlando, FL 32804, USA
| | - Sabrina Pavri
- Division of Plastic Surgery, Department of Surgery, AdventHealth Hospital, Orlando, FL 32804, USA;
| |
Collapse
|
3
|
Moore HN, Goncalves MD, Johnston AM, Mayer EL, Rugo HS, Gradishar WJ, Zylla DM, Bergenstal RM. Effective Strategies for the Prevention and Mitigation of Phosphatidylinositol-3-Kinase Inhibitor-Associated Hyperglycemia: Optimizing Patient Care. Clin Breast Cancer 2025; 25:1-11. [PMID: 39462728 DOI: 10.1016/j.clbc.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024]
Abstract
Hyperglycemia is a common adverse event (AE) associated with phosphatidylinositol-3-kinase inhibitors (PI3Kis) and considered an on-target effect. Presence of hyperglycemia is associated with poor outcomes in patients with cancer, and there is need for further refinement of hyperglycemia prevention and mitigation strategies in patients receiving PI3Kis. In this review, the authors highlight effective strategies for preventing PI3Ki-induced hyperglycemia before and during treatment as well as hyperglycemia management. Prior to initiating treatment with PI3Ki, identify baseline risk factors of patients at increased risk for developing hyperglycemia, which include older age, obesity, and glycosylated hemoglobin (HbA1c) 5.7%-6.4% (prediabetes or Type 2 diabetes). To prevent new-onset hyperglycemia, optimize blood glucose, and recommend a low-carbohydrate (60-130 g/day) diet along with regular exercise to all patients prior to initiating the PI3Ki. Prophylactic metformin may be considered in all patients starting a PI3Ki with HbA1c ≤6.4%. Although existing recommendations support monitoring fasting blood glucose (FBG) once weekly (twice-weekly for intermediate-risk, daily for high-risk patients) and HbA1c every 3 months upon initiation of PI3Ki, more frequent FBG monitoring may be considered for prompt detection of hyperglycemia. Experts also recommend considering postprandial glucose monitoring because it is an early indicator of glucose intolerance. If hyperglycemia develops, metformin (first-line) and/or sodium glucose co-transporter 2 inhibitors or thiazolidinediones (second-/third-line) are the preferred agents; consider early referral to an endocrinologist. In conclusion, hyperglycemia is a common but manageable AE associated with PI3Kis. Multidisciplinary approach to the prevention, monitoring, and management of hyperglycemia optimizes patient care and allows patients to maintain therapy on PI3Ki.
Collapse
Affiliation(s)
| | | | | | - Erica L Mayer
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Hope S Rugo
- Department of Medicine (Hematology/Oncology), University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | | | - Dylan M Zylla
- The Cancer Research Center, HealthPartners Institute, Minneapolis, MN
| | | |
Collapse
|
4
|
Jhaveri KL, Lim E, Jeselsohn R, Ma CX, Hamilton EP, Osborne C, Bhave M, Kaufman PA, Beck JT, Manso Sanchez L, Parajuli R, Wang HC, Tao JJ, Im SA, Harnden K, Yonemori K, Dhakal A, Neven P, Aftimos P, Pierga JY, Lu YS, Larson T, Jerez Y, Sideras K, Sohn J, Kim SB, Saura C, Bardia A, Sammons SL, Bacchion F, Li Y, Yuen E, Estrem ST, Rodrik-Outmezguine V, Nguyen B, Ismail-Khan R, Smyth L, Beeram M. Imlunestrant, an Oral Selective Estrogen Receptor Degrader, as Monotherapy and in Combination With Targeted Therapy in Estrogen Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: Phase Ia/Ib EMBER Study. J Clin Oncol 2024; 42:4173-4186. [PMID: 39241211 PMCID: PMC11637582 DOI: 10.1200/jco.23.02733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/08/2024] Open
Abstract
PURPOSE Imlunestrant is a next-generation oral selective estrogen receptor (ER) degrader designed to deliver continuous ER target inhibition, including in ESR1-mutant breast cancer. This phase Ia/b trial determined the recommended phase II dose (RP2D), safety, pharmacokinetics, and efficacy of imlunestrant, as monotherapy and in combination with targeted therapy, in ER-positive (ER+) advanced breast cancer (ABC) and endometrial endometrioid cancer. The ER+/human epidermal growth factor receptor 2-negative (HER2-) ABC experience is reported here. METHODS An i3+3 dose-escalation design was used, followed by dose expansions of imlunestrant as monotherapy or in combination with abemaciclib with or without aromatase inhibitor (AI), everolimus, or alpelisib. Imlunestrant was administered orally once daily and with the combination partner per label. RESULTS Overall, 262 patients with ER+/HER2- ABC were treated (phase Ia, n = 74; phase Ib, n = 188). Among patients who received imlunestrant monotherapy (n = 114), no dose-limiting toxicities or discontinuations occurred. At the RP2D (400 mg once daily), patients (n = 51) reported grade 1-2 nausea (39.2%), fatigue (39.2%), and diarrhea (29.4%). Patients at RP2D had received previous cyclin-dependent kinase 4/6 inhibitor (CDK4/6i; 92.2%), fulvestrant (41.2%), and chemotherapy (29.4%) for ABC and achieved a median progression-free survival (mPFS) of 7.2 months (95% CI, 3.7 to 8.3). Among patients who received imlunestrant + abemaciclib (n = 42) and imlunestrant + abemaciclib + AI (n = 43), most (69.4%) were treatment-naïve for ABC; all were CDK4/6i-naïve. Patients treated with imlunestrant + everolimus (n = 42)/alpelisib (n = 21) had received previous CDK4/6i (100%), fulvestrant (34.9%), and chemotherapy (17.5%) for ABC. No new safety signals or interactions with partnered drugs were observed. The mPFS was 19.2 months (95% CI, 13.8 to not available) for imlunestrant + abemaciclib and was not reached for imlunestrant + abemaciclib + AI. The mPFS with imlunestrant + everolimus/alpelisib was 15.9 months (95% CI, 11.3 to 19.1)/9.2 months (95% CI, 3.7 to 11.1). Antitumor activity was evident regardless of ESR1 mutation status. CONCLUSION Imlunestrant, as monotherapy or in combination with targeted therapy, had a manageable safety profile with evidence of preliminary antitumor activity in ER+/HER2- ABC.
Collapse
Affiliation(s)
- Komal L. Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Elgene Lim
- Garvan Institute of Medical Research, St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | | | - Cynthia X. Ma
- Washington University School of Medicine, St Louis, MO
| | | | - Cynthia Osborne
- US Oncology Research, McKesson Specialty Health, The Woodlands, TX
- Texas Oncology, Baylor-Sammons Cancer Center, Dallas, TX
| | | | | | | | | | | | | | - Jessica J. Tao
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | | | | | - Ajay Dhakal
- University of Rochester Medical Center, Rochester, NY
| | - Patrick Neven
- Universitaire Ziekenhuizen-Leuven Cancer Institute, Leuven, Belgium
| | - Philippe Aftimos
- Université Libre de Bruxelles, Hôpital Universitaire de Bruxelles, Institut Jules Bordet, Brussels, Belgium
| | | | - Yen-Shen Lu
- National Taiwan University Hospital, Taipei, Taiwan
| | - Timothy Larson
- Minnesota Oncology/The US Oncology Network, Minneapolis, MN
| | - Yolanda Jerez
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERONC, Madrid, Spain
| | | | - Joohyuk Sohn
- Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Cristina Saura
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Sarah L. Sammons
- Dana-Farber Cancer Institute, Boston, MA
- Duke Cancer Institute, Duke University, Durham, NC
| | | | - Yujia Li
- Eli Lilly and Company, Indianapolis, IN
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Fanucci K, Giordano A, Erick T, Tolaney SM, Sammons S. Practical treatment strategies and novel therapies in the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway in hormone receptor-positive/human epidermal growth factor receptor 2 (HER2)-negative (HR+/HER2-) advanced breast cancer. ESMO Open 2024; 9:103997. [PMID: 39674130 PMCID: PMC11699375 DOI: 10.1016/j.esmoop.2024.103997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 12/16/2024] Open
Abstract
Mutations in the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway occur in 30%-40% of patients with advanced hormone receptor-positive/human epidermal growth factor receptor 2 (HER2)-negative (HR+/HER2-) breast cancer. For most patients, endocrine therapy with a cyclin-dependent kinase 4/6 (CDK4/6) inhibitor is the first-line treatment. Recent studies indicate that adding inavolisib, a PI3Kα inhibitor, to palbociclib/fulvestrant benefits patients with endocrine-resistant HR+/HER2- metastatic breast cancer with a PIK3CA mutation. Alpelisib and capivasertib are both US Food and Drug Administration (FDA) approved in combination with fulvestrant in patients with endocrine-resistant HR+/HER2-, PIK3CA-mutant metastatic breast cancer, both with activity in the post-CDK4/6 setting. Capivasertib added to fulvestrant is the first AKT inhibitor to show a significant progression-free survival benefit with a trend for overall survival benefit and the only approved option for patients with phosphate and tensin homolog (PTEN) or AKT alterations. Toxicity profiles of all agents necessitate careful patient selection. Several mutant-selective and pan-mutant-selective novel inhibitors are under investigation with the potential to improve tolerability and efficacy.
Collapse
Affiliation(s)
- K Fanucci
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, USA; Harvard Medical School, Boston, USA. https://twitter.com/KristinaFanucci
| | - A Giordano
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, USA; Harvard Medical School, Boston, USA. https://twitter.com/antgiorda
| | - T Erick
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, USA. https://twitter.com/DrTimothyErick
| | - S M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, USA; Harvard Medical School, Boston, USA. https://twitter.com/stolaney1
| | - S Sammons
- Medical Oncology, Dana-Farber Cancer Institute, Boston, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, USA; Harvard Medical School, Boston, USA.
| |
Collapse
|
6
|
Layman RM, Han HS, Rugo HS, Stringer-Reasor EM, Specht JM, Dees EC, Kabos P, Suzuki S, Mutka SC, Sullivan BF, Gorbatchevsky I, Wesolowski R. Gedatolisib in combination with palbociclib and endocrine therapy in women with hormone receptor-positive, HER2-negative advanced breast cancer: results from the dose expansion groups of an open-label, phase 1b study. Lancet Oncol 2024; 25:474-487. [PMID: 38547892 DOI: 10.1016/s1470-2045(24)00034-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND The PI3K-mTOR pathway is frequently dysregulated in breast cancer. Combining an inhibitor targeting all class I PI3K isoforms and mTOR complex 1 (mTORC1)-mTOR complex 2 (mTORC2) with endocrine therapy and a CDK4/6 inhibitor might provide more effective tumour control than standard-of-care therapy. To evaluate this hypothesis, gedatolisib, a pan-PI3K-mTOR inhibitor, was assessed in a phase 1b trial combined with palbociclib and endocrine therapy in patients with hormone receptor-positive, HER2-negative, advanced breast cancer. Results from the dose expansion portion of this trial are reported herein. METHODS This multicentre, open-label, phase 1b study recruited female patients aged at least 18 years from 17 sites across the USA with hormone-receptor-positive, HER2-negative, advanced breast cancer and an Eastern Cooperative Oncology Group performance status of 0-1. Four patient groups were studied in the dose expansion portion of the study: treatment-naive in the advanced setting (first line; group A), progression on 1-2 lines of endocrine therapy but CDK4/6 inhibitor-naive (group B); and one or more previous lines (second-line and higher) of therapy, including a CDK4/6 inhibitor (groups C and D). Gedatolisib 180 mg was administered intravenously weekly in 28-day treatment cycles for groups A-C, and on days 1, 8, and 15 for group D. Letrozole (group A), fulvestrant (groups B-D), and palbociclib (all groups) were administered at standard doses and schedules. The primary endpoint was investigator-assessed objective response rate per RECIST version 1.1 in the evaluable analysis set. This trial is completed and registered with ClinicalTrials.gov, NCT02684032. FINDINGS Between Dec 19, 2017, and June 19, 2019, 103 female participants were enrolled in the dose expansion groups A (n=31), B (n=13), C (n=32), and D (n=27). Median follow-up was 16·6 months (IQR 5·7-48·4) for group A, 11·0 months (7·6-16·9) for group B, 3·6 months (1·8-7·5) for group C, and 9·4 months (5·3-16·7) for group D for the primary endpoint. Gedatolisib, palbociclib, and endocrine therapy induced an objective response in 23 (85·2%; 90% CI 69·2-94·8) of 27 evaluable first-line participants (group A). In the second-line and higher setting, an objective response was observed in eight (61·5%; 90% CI 35·5-83·4) of 13 evaluable group B participants, seven (25·0%; 12·4-41·9) of 28 evaluable group C participants, and 15 (55·6%; 38·2-72·0) of 27 evaluable group D participants; this included participants with both wild-type and mutated PIK3CA tumours. The most common grade 3-4 treatment-related adverse events were neutropenia (65 [63%] of 103), stomatitis (28 [27%]), and rash (21 [20%]). Grade 3-4 hyperglycaemia was reported in six (6%) participants. 23 (22%) of 103 participants had a treatment-related serious adverse event, and there were no treatment-related deaths. Nine (9%) participants discontinued treatment because of a treatment-emergent adverse event. INTERPRETATION Gedatolisib plus palbociclib and endocrine therapy showed a promising objective response rate compared with the published results for standard-of-care therapies and had an acceptable safety profile. FUNDING Pfizer and Celcuity.
Collapse
Affiliation(s)
- Rachel M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Hyo S Han
- Moffit Cancer Center, Tampa, FL, USA
| | - Hope S Rugo
- Division of Hematology and Oncology, University of California, San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | - Erica M Stringer-Reasor
- Division of Hematology Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer M Specht
- Division of Hematology and Oncology, Fred Hutch Cancer Center, University of Washington, Seattle, WA, USA
| | - E Claire Dees
- Division of Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Peter Kabos
- Division of Medical Oncology, University of Colorado Hospital, Aurora, CO, USA
| | | | | | | | | | - Robert Wesolowski
- Department. of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
7
|
AlJabban A, Paik H, Aster JC, Berliner N, Brouillard J, Brown JR, Burns KH, Castillo JJ, Card J, Dal Cin P, DeAngelo DJ, Dorfman DM, Ebert BL, Garcia JS, Jacobson CA, Lakhani H, Laubach JP, Ligon AH, Lindeman NI, Lindsley RC, Lovitch SB, Luskin MR, Morgan EA, Nowak A, Petrides A, Pinkus GS, Pozdnyakova O, Steensma DP, Stone RM, Weinberg OK, Winer ES, Kim AS. Optimization of Advanced Molecular Genetic Testing Utilization in Hematopathology: A Goldilocks Approach to Bone Marrow Testing. JCO Oncol Pract 2024; 20:220-227. [PMID: 37683132 DOI: 10.1200/op.23.00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 09/10/2023] Open
Abstract
PURPOSE This study investigated the effectiveness of algorithmic testing in hematopathology at the Brigham and Women's Hospital and Dana-Farber Cancer Institute (DFCI). The algorithm was predicated on test selection after an initial pathologic evaluation to maximize cost-effective testing, especially for expensive molecular and cytogenetic assays. MATERIALS AND METHODS Standard ordering protocols (SOPs) for 17 disease categories were developed and encoded in a decision support application. Six months of retrospective data from application beta testing was obtained and compared with actual testing practices during that timeframe. In addition, 2 years of prospective data were also obtained from patients at one community satellite site. RESULTS A total of 460 retrospective cases (before introduction of algorithmic testing) and 109 prospective cases (following introduction) were analyzed. In the retrospective data, 61.7% of tests (509 of 825) were concordant with the SOPs while 38.3% (316 of 825) were overordered and 30.8% (227 of 736) of SOP-recommended tests were omitted. In the prospective data, 98.8% of testing was concordant (244 of 247 total tests) with only 1.2% overordered tests (3 of 247) and 7.6% omitted tests (20 of 264 SOP-recommended tests; overall P < .001). The cost of overordered tests before implementing SOP indicates a potential annualized saving of $1,347,520 in US dollars (USD) in overordered testing at Brigham and Women's Hospital/DFCI. Only two of 316 overordered tests (0.6%) returned any additional information, both for extremely rare clinical circumstances. CONCLUSION Implementation of SOPs dramatically improved test ordering practices, with a just right number of ancillary tests that minimizes cost and has no significant impact on acquiring key informative test results.
Collapse
Affiliation(s)
- Ali AlJabban
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Henry Paik
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Enterprise Research IS (ERIS), Digital, Mass General Brigham, Boston, MA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nancy Berliner
- Harvard Medical School, Boston, MA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA
| | | | - Jennifer R Brown
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kathleen H Burns
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Jorge J Castillo
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - James Card
- Department of Quality and Safety, Dana-Farber Cancer Institute, Boston, MA
| | - Paola Dal Cin
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Daniel J DeAngelo
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Benjamin L Ebert
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jacqueline S Garcia
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Caron A Jacobson
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Hakim Lakhani
- Department of Quality and Safety, Dana-Farber Cancer Institute, Boston, MA
| | - Jacob P Laubach
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Azra H Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Neal I Lindeman
- Department of Pathology, Weill Cornell Medicine, New York, NY
| | - R Coleman Lindsley
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Scott B Lovitch
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Marlise R Luskin
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Elizabeth A Morgan
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Andrew Nowak
- Department of Performance Improvement, Stanford Medicine Children's Health, Stanford, CA
| | - Athena Petrides
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Enterprise Research IS (ERIS), Digital, Mass General Brigham, Boston, MA
| | - Geraldine S Pinkus
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Olga Pozdnyakova
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - David P Steensma
- Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA
| | - Richard M Stone
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Olga K Weinberg
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Eric S Winer
- Harvard Medical School, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
8
|
Jauslin WT, Schild M, Schaefer T, Borsari C, Orbegozo C, Bissegger L, Zhanybekova S, Ritz D, Schmidt A, Wymann M, Gillingham D. A high affinity pan-PI3K binding module supports selective targeted protein degradation of PI3Kα. Chem Sci 2024; 15:683-691. [PMID: 38179525 PMCID: PMC10763047 DOI: 10.1039/d3sc04629j] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/01/2023] [Indexed: 01/06/2024] Open
Abstract
Class I phosphoinositide 3-kinases (PI3Ks) control cellular growth, but are also essential in insulin signaling and glucose homeostasis. Pan-PI3K inhibitors thus generate substantial adverse effects, a reality that has plagued drug development against this target class. We present here evidence that a high affinity binding module with the capacity to target all class I PI3K isoforms can facilitate selective degradation of the most frequently mutated class I isoform, PI3Kα, when incorporated into a cereblon-targeted (CRBN) degrader. A systematic proteomics study guided the fine tuning of molecular features to optimize degrader selectivity and potency. Our work resulted in the creation of WJ112-14, a PI3Kα-specific nanomolar degrader that should serve as an important research tool for studying PI3K biology. Given the toxicities observed in the clinic with unselective PI3Kα inhibitors, the results here offer a new approach toward selectively targeting this frequently mutated oncogenic driver.
Collapse
Affiliation(s)
| | - Matthias Schild
- Department of Chemistry, University of Basel 4056 Basel Switzerland
| | - Thorsten Schaefer
- Department of Biomedicine, University of Basel 4031 Basel Switzerland
| | - Chiara Borsari
- Department of Biomedicine, University of Basel 4031 Basel Switzerland
| | - Clara Orbegozo
- Department of Biomedicine, University of Basel 4031 Basel Switzerland
| | - Lukas Bissegger
- Department of Biomedicine, University of Basel 4031 Basel Switzerland
| | | | - Danilo Ritz
- Proteomics Core Facility, Biozentrum, University of Basel 4056 Basel Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel 4056 Basel Switzerland
| | - Matthias Wymann
- Department of Biomedicine, University of Basel 4031 Basel Switzerland
| | | |
Collapse
|
9
|
Shen S, Chen Y, Carpio A, Chang C, Iyengar NM. Incidence, risk factors, and management of alpelisib-associated hyperglycemia in metastatic breast cancer. Cancer 2023; 129:3854-3861. [PMID: 37743730 PMCID: PMC10863751 DOI: 10.1002/cncr.34928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/03/2023] [Accepted: 05/24/2023] [Indexed: 09/26/2023]
Abstract
PURPOSE The combination of fulvestrant with alpelisib, a PI3K inhibitor, improves progression-free survival in metastatic hormone receptor-positive, PIK3CA-mutant breast cancer. This study describes the incidence, risk factors, and treatment of alpelisib-associated hyperglycemia. METHODS Patients with metastatic breast cancer who received alpelisib from 2013 to 2021 at Memorial Sloan Kettering Cancer Center were included in this retrospective study. Alpelisib prescription dates and patient/tumor characteristics were abstracted from medical records. Risk factors associated with hyperglycemia and alpelisib dose reduction/discontinuation were evaluated using Pearson's χ2 tests. RESULTS Among 247 patients, baseline median body mass index was 25.4 kg/m2 and median hemoglobin A1c (HbA1c) was 5.5%. A total of 152 patients (61.5%) developed any-grade hyperglycemia and 72 patients (29.2%) developed grade 3-4 hyperglycemia; median time to onset was 16 days. A total of 100 patients (40.5%) received alpelisib on a clinical trial; rates of hyperglycemia were significantly higher in patients treated as standard care versus on a clinical trial (any-grade hyperglycemia 80.3% vs. 34.0%, grade 3-4 hyperglycemia 40.2% vs. 13.0%, p < .001). Baseline HbA1c was significantly associated with development of hyperglycemia (p < .001) and alpelisib dose reduction/discontinuation (p = .015). Among those who developed hyperglycemia, 101 (40.9%) received treatment, most commonly with metformin. A total of 49 patients (19.8%) were referred to an endocrinologist, which was associated with SGLT2 inhibitor prescription (p = .007). CONCLUSIONS Rates of hyperglycemia among patients treated with alpelisib as standard care were significantly higher than patients treated on clinical trials. Elevated baseline HbA1c is associated with alpelisib-induced hyperglycemia and requiring dose modification. Optimization of glycemic status before alpelisib initiation should become routine practice.
Collapse
Affiliation(s)
- Sherry Shen
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yuan Chen
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrea Carpio
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Neil M. Iyengar
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical Center, New York, NY
| |
Collapse
|
10
|
Buckbinder L, St. Jean DJ, Tieu T, Ladd B, Hilbert B, Wang W, Alltucker JT, Manimala S, Kryukov GV, Brooijmans N, Dowdell G, Jonsson P, Huff M, Guzman-Perez A, Jackson EL, Goncalves MD, Stuart DD. STX-478, a Mutant-Selective, Allosteric PI3Kα Inhibitor Spares Metabolic Dysfunction and Improves Therapeutic Response in PI3Kα-Mutant Xenografts. Cancer Discov 2023; 13:2432-2447. [PMID: 37623743 PMCID: PMC10618743 DOI: 10.1158/2159-8290.cd-23-0396] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/24/2023] [Accepted: 08/23/2023] [Indexed: 08/26/2023]
Abstract
Phosphoinositide 3-kinase α (PIK3CA) is one of the most mutated genes across cancers, especially breast, gynecologic, and head and neck squamous cell carcinoma tumors. Mutations occur throughout the gene, but hotspot mutations in the helical and kinase domains predominate. The therapeutic benefit of isoform-selective PI3Kα inhibition was established with alpelisib, which displays equipotent activity against the wild-type and mutant enzyme. Inhibition of wild-type PI3Kα is associated with severe hyperglycemia and rash, which limits alpelisib use and suggests that selectively targeting mutant PI3Kα could reduce toxicity and improve efficacy. Here we describe STX-478, an allosteric PI3Kα inhibitor that selectively targets prevalent PI3Kα helical- and kinase-domain mutant tumors. STX-478 demonstrated robust efficacy in human tumor xenografts without causing the metabolic dysfunction observed with alpelisib. Combining STX-478 with fulvestrant and/or cyclin-dependent kinase 4/6 inhibitors was well tolerated and provided robust and durable tumor regression in ER+HER2- xenograft tumor models. SIGNIFICANCE These preclinical data demonstrate that the mutant-selective, allosteric PI3Kα inhibitor STX-478 provides robust efficacy while avoiding the metabolic dysfunction associated with the nonselective inhibitor alpelisib. Our results support the ongoing clinical evaluation of STX-478 in PI3Kα-mutated cancers, which is expected to expand the therapeutic window and mitigate counterregulatory insulin release. See related commentary by Kearney and Vasan, p. 2313. This article is featured in Selected Articles from This Issue, p. 2293.
Collapse
Affiliation(s)
| | - David J. St. Jean
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | - Trang Tieu
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | - Brendon Ladd
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | - Brendan Hilbert
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | - Weixue Wang
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | | | - Samantha Manimala
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | | | | | - Gregory Dowdell
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | - Philip Jonsson
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | - Michael Huff
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| | | | - Erica L. Jackson
- Department of Biology, Scorpion Therapeutics, South San Francisco, California
| | - Marcus D. Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Darrin D. Stuart
- Research and Development, Scorpion Therapeutics, Boston, Massachusetts
| |
Collapse
|
11
|
LeVee A, Mortimer J. The Challenges of Treating Patients with Breast Cancer and Obesity. Cancers (Basel) 2023; 15:2526. [PMID: 37173991 PMCID: PMC10177120 DOI: 10.3390/cancers15092526] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Obesity is defined as a body mass index (BMI) of 30 kg/m2 or more and is associated with worse outcomes in patients with breast cancer, resulting in an increased incidence of breast cancer, recurrence, and death. The incidence of obesity is increasing, with almost half of all individuals in the United States classified as obese. Patients with obesity present with unique pharmacokinetics and physiology and are at increased risk of developing diabetes mellitus and cardiovascular disease, which leads to specific challenges when treating these patients. The aim of this review is to summarize the impact of obesity on the efficacy and toxicity of systemic therapies used for breast cancer patients, describe the molecular mechanisms through which obesity can affect systemic therapies, outline the existing American Society of Clinical Oncology (ASCO) guidelines for treating patients with cancer and obesity, and highlight additional clinical considerations for treating patients with obesity and breast cancer. We conclude that further research on the biological mechanisms underlying the obesity-breast cancer link may offer new treatment strategies, and clinicals trials that focus on the treatment and outcomes of patients with obesity and all stages of breast cancer are needed to inform future treatment guidelines.
Collapse
Affiliation(s)
- Alexis LeVee
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | | |
Collapse
|
12
|
Wang Y, Ma Z, An Z, Zhang Y, Feng X, Yu X. Risk of cutaneous adverse events in cancer patients treated with phosphatidylinositol-3-kinase inhibitors: A systematic review and meta-analysis of randomized controlled trials. Cancer Med 2022; 12:2227-2237. [PMID: 35986570 PMCID: PMC9939201 DOI: 10.1002/cam4.5153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cutaneous adverse effects (AEs) are common following the phosphoinositide-3-kinase (PI3K) inhibitors treatment. We aim to estimate the incidence and risk of PI3K inhibitor-related cutaneous AEs. METHODS The protocol was submitted to the PROSPERO registry. We searched ClinicalTrials.gov and international databases up to July 29, 2022. Meta-analysis was conducted by using risk ratios (RRs) with 95% confidence intervals (CIs). RESULTS Fourteen randomized controlled trials (RCTs) comprising 3877 patients were analyzed in this study. Compared with control arms, PI3K inhibitors showed a significant increase in the risk of all-grade rash, high-grade rash, and serious rash events (RR 2.29, 95% CI 1.58-3.31, p < 0.00001; RR 9.34, 95% CI 4.21-20.69, p < 0.00001; RR 5.11, 95% CI 2.11-12.36, p = 0.0003). The overall incidences of all-grade rash and high-grade rash were 26.2% (592/2257) and 4.4% (66/1487). Subgroup analyses of all-grade rash according to cancer types and PI3K inhibitor assignations identified the significant associations. PI3K inhibitors also significantly increased the risk of pruritus and dry skin (RR 1.63, 95% CI 1.14-2.33, p = 0.007; RR 3.34, 95% CI 2.30-4.85, p < 0.00001), with incidences of 13.4% (284/2115) and 9.8% (141/1436) in the treatment group. CONCLUSION There is a significantly increased risk of some cutaneous AEs in patients using PI3K inhibitors. Advance intervention is recommended in case of severe and life-threatening events. Further research is required to investigate the risk factors and pathogenesis.
Collapse
Affiliation(s)
- Yushu Wang
- Department of PharmacyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina,Department of PharmacyBeijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care HospitalBeijingChina
| | - Zhuo Ma
- Department of PharmacyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Zhuoling An
- Department of PharmacyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Yi Zhang
- Department of PharmacyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina,Department of PharmacyBeijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care HospitalBeijingChina
| | - Xin Feng
- Department of PharmacyBeijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care HospitalBeijingChina
| | - Xiaojia Yu
- Department of PharmacyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
13
|
Jacobs AT, Martinez Castaneda-Cruz D, Rose MM, Connelly L. Targeted therapy for breast cancer: An overview of drug classes and outcomes. Biochem Pharmacol 2022; 204:115209. [PMID: 35973582 DOI: 10.1016/j.bcp.2022.115209] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 12/20/2022]
Abstract
The last 25 years have seen significant growth in new therapeutic options for breast cancer, termed targeted therapies based on their ability to block specific pathways known to drive breast tumor growth and survival. Introduction of these drugs has been made possible through advances in the understanding of breast cancer biology. While the promise of targeted therapy for breast cancer has been clear for some time, the experience of the clinical use of multiple drugs and drug classes allows us to now present a summary and perspective as to the success and impact of this endeavor. Here we will review breast cancer targeted therapeutics in clinical use. We will provide the rationale for their indications and summarize clinical data in patients with different breast cancer subtypes, their impact on breast cancer progression and survival and their major adverse effects. The focus of this review will be on the development that has occurred within classes of targeted therapies and subsequent impact on breast cancer patient outcomes. We will conclude with a perspective on the role of targeted therapy in breast cancer treatment and highlight future areas of development.
Collapse
Affiliation(s)
- Aaron T Jacobs
- California University of Science and Medicine, 1501 Violet Street, Colton, CA 92324, United States
| | | | - Mark M Rose
- California University of Science and Medicine, 1501 Violet Street, Colton, CA 92324, United States
| | - Linda Connelly
- California University of Science and Medicine, 1501 Violet Street, Colton, CA 92324, United States.
| |
Collapse
|
14
|
Expert consensus on the clinical application of PI3K/AKT/mTOR inhibitors in the treatment of advanced breast cancer. CANCER INNOVATION 2022; 1:25-54. [PMID: 38089455 PMCID: PMC10686175 DOI: 10.1002/cai2.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 04/07/2024]
Abstract
Phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB or AKT)/mammalian target of rapamycin (mTOR) signaling pathway (PAM pathway) plays an important role in the development of breast cancer and are closely associated with the resistance to endocrine therapy in advanced breast cancer. Therefore, anticancer treatment targeting key molecules in this signaling pathway has become a research hotspot in recent years. Randomized clinical trials have demonstrated that PI3K/AKT/mTOR inhibitors bring significant clinical benefit to patients with advanced breast cancer, especially to those with hormone receptor (HR)-positive, human epidermal growth factor receptor (HER) 2-negative advanced breast cancer. Alpelisib, a PI3K inhibitor, and everolimus, an mTOR inhibitor, have been approved by FDA. Based on their high efficacy and relatively good safety profile, an expanded indication of everolimus in breast cancer has been approved by National Medical Products Administration (NMPA). Alpelisib is expected to be approved in China in the near future. The members of the consensus expert panel reached this consensus to comprehensively define the role of PI3K/AKT/mTOR signaling pathway in breast cancer, efficacy and clinical applications of PI3K/AKT/mTOR inhibitors, management of adverse reactions, and PIK3CA mutation detection, to promote the understanding of PI3K/AKT/mTOR inhibitors for Chinese oncologists, improve clinical decision-making, and prolong the survival of target patient population.
Collapse
|
15
|
Miller J, Armgardt E, Svoboda A. The efficacy and safety of alpelisib in breast cancer: A real-world analysis. J Oncol Pharm Pract 2022:10781552221096413. [PMID: 35450470 DOI: 10.1177/10781552221096413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Published trials of alpelisib + fulvestrant demonstrate efficacy and high rates of adverse effects as a first-line treatment option for metastatic breast cancer and as an option after cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i). The purpose of this analysis is to determine the real-world efficacy and safety of this regimen in heavily pretreated patients. This is a retrospective cohort analysis evaluating patients receiving alpelisib + fulvestrant for hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) metastatic breast cancer who previously received ≤ 2 lines of therapy in the metastatic setting and those who previously received ≥ 3 lines of therapy in the metastatic setting. Adverse effects, specifically hyperglycemia, rash, and diarrhea, were reported for the entire population. Thirty-three patients were included in this analysis. Progression-free survival (PFS), overall survival, time to change in therapy, and time to discontinuation were similar in the two groups. Forty-nine percent of patients discontinued alpelisib + fulvestrant due to progression of disease, and 27% of patients discontinued treatment due to adverse effects. Hyperglycemia, rash, and diarrhea occurred at high rates: 66.7%, 45.5%, and 72.7%, respectively. All three of these adverse effects required hospitalizations and pharmacological treatment. This analysis demonstrates that the outcomes of alpelisib + fulvestrant were worse in the real-world salvage setting in HR+, HER2- metastatic breast cancer as compared to the front-line setting. The real-world tolerability of this regimen is still of great concern.
Collapse
Affiliation(s)
- Jenn Miller
- 24560Northwestern Memorial Hospital, Chicago, IL, USA
| | - Emily Armgardt
- 88966Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Alison Svoboda
- 88966Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|