1
|
Zhang Y, Xu X, Yang K, Wang S, Zhang T, Hui F, Zheng F, Geng H, Xu C, Xun F, Xu Z, Wang C, Hou S, Song A, Ren T, Zhao Q. The efficacy and safety of PI3K and AKT inhibitors for patients with cancer: A systematic review and network meta-analysis. Eur J Pharmacol 2024; 983:176952. [PMID: 39216745 DOI: 10.1016/j.ejphar.2024.176952] [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: 07/13/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Inhibiting PI3K/AKT pathway activation may hinder the occurrence and progression of cancer. The aim of this study was to evaluate the efficacy and safety of the PI3K/AKT inhibitors and determine the most appropriate inhibitor for different cancer types. METHODS Electronic databases up to June 2024 were used to examine the efficacy and safety of PI3K inhibitors (alpelisib, copanlisib, duvelisib, and idelalisib) and AKT inhibitors (capivasertib, ipatasertib and MK-2206) for the treatment of cancer. Data was assessed with a random-effect pairwise and network meta-analysis. Randomized controlled trials and retrospective studies were eligible if they compared PI3K or AKT inhibitors with non-PI3K/AKT controls with no restriction. RESULTS The results were based on 34 studies from 34 published articles and 6 online registration trials (6710 patients). According to pairwise meta-analysis, PI3K/AKT inhibitors showed to be highly effective, especially for treating mutant cancers, but had poor safety profiles. According to our network meta-analysis, PI3K/AKT inhibitors, especially the AKT inhibitor capivasertib, are effective for treating solid cancers such as breast cancer (BC). Moreover, PI3K inhibitors, especially idelalisib, were effective for treating hematologic cancers such as chronic lymphocytic leukemia (CLL). CONCLUSIONS The PI3K/AKT inhibitors are effective in patients with genetic mutations. For solid cancers such as BC, capivasertib was efficacy and safety. For hematological cancers represented by CLL, idelalisib was efficacy and safety. The above studies can be used when recommending appropriate targeted therapies for patients with different cancer types.
Collapse
Affiliation(s)
- Yingshi Zhang
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Xiangbo Xu
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Kaisi Yang
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Shuai Wang
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Tianqi Zhang
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Fuhai Hui
- Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Fangyuan Zheng
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Hefeng Geng
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Chang Xu
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Fanghua Xun
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; General Hospital of Northern Theater Command, China Medical University, Shenyang city, Liaoning province, PR China.
| | - Ziang Xu
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Chengkang Wang
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Shanbo Hou
- Luoxin Pharmaceuticals Group Stock Co., Ltd., Linyi, PR China.
| | - Aigang Song
- Luoxin Pharmaceuticals Group Stock Co., Ltd., Linyi, PR China.
| | - Tianshu Ren
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| | - Qingchun Zhao
- Teaching hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, 100016, Shenyang city, Liaoning province, PR China; Department of Clinical Pharmacy, Shenyang Pharmaceutical University, Shenyang city, Liaoning province, PR China.
| |
Collapse
|
2
|
Lin X, Zhang Y, Huang H, Zhuang W, Wu L. Post-marketing safety concern of PI3K inhibitors in the cancer therapies: an 8-year disproportionality analysis from the FDA Adverse Event Reporting System. Expert Opin Drug Saf 2024:1-12. [PMID: 39083397 DOI: 10.1080/14740338.2024.2387317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/18/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND The Phosphoinositide 3-kinases (PI3Ks) family plays a crucial role in tumorigenesis. Alpelisib (inhibiting PI3Kα), copanlisib (inhibiting PI3Kα andPI3Kδ), duvelisib (inhibiting PI3Kδ and PI3Kγ), and idelalisib (inhibiting PI3Kδ) were developed to target the PI3K pathway. However, the toxicity limits their application to some extent. It's necessary to investigate the adverse effects (AEs) of these inhibitors. RESEARCH DESIGN AND METHODS We conducted a comparative analysis of the safety signals of AEs in PI3K inhibitors using disproportionality analysis in the FDA Adverse Event Reporting System database(FAERS). RESULTS Our study identified significant safety signals for metabolic disorders with all PI3K inhibitors. Notable safety signals for gastrointestinal disorders were observed with most PI3K inhibitors, with the exception of copanlisib. Common AEs shared among all PI3K inhibitors included colitis and dehydration. Alpelisib displayed unique AEs associated with metabolic disorders, whereas copanlisib exhibited idiosyncratic AEs linked to cardiac and vascular disorders. Stevens-Johnson syndrome emerged as a common severe adverse event (SAE) among alpelisib, copanlisib, and idelalisib, while febrile neutropenia was prevalent among copanlisib, duvelisib, and idelalisib. Intestinal perforation was solely associated with alpelisib. CONCLUSIONS The safety profiles of the five PI3K inhibitors vary concerning adverse events. These findings could guide drug selection and inform future prospective research.
Collapse
Affiliation(s)
- Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, Guangdong Province, China
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong Province, China
| | - Yimin Zhang
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong Province, China
| | - Hongyan Huang
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Zhuang
- Department of Pharmacy, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian Province, China
| | - Lisha Wu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Oncology, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
| |
Collapse
|
3
|
Sirico M, Jacobs F, Molinelli C, Nader-Marta G, Debien V, Dewhurst HF, Palleschi M, Merloni F, Gianni C, De Giorgi U, de Azambuja E. Navigating the complexity of PI3K/AKT pathway in HER-2 negative breast cancer: biomarkers and beyond. Crit Rev Oncol Hematol 2024; 200:104404. [PMID: 38815877 DOI: 10.1016/j.critrevonc.2024.104404] [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: 12/21/2023] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024] Open
Abstract
The results of the SOLAR-1 and CAPItello-291, highlight the benefit of the ɑ-selective phosphoinositide 3-Kinase Pathway inhibitor (PI3Ki) alpelisib and the AKT inhibitor (AKTi) capivasertib in patients with hormone receptor-positive (HR+)/Human Epidermal Growth Factor Receptor 2 (HER2)- negative metastatic breast cancer (mBC) that have PIK3CA/AKT1/PTEN tumour alterations. Although effective, these drugs are associated with significant toxicities, which often limit their use, particularly in frail patients. Following the recent incorporation of these agents into clinical practice, and with many others currently in development, significant challenges have emerged, particularly those regarding biomarkers for patient selection. This review will discuss biomarkers of response and their resistance to PI3K/AKT inhibitors (PI3K/AKTis) in HR+/HER- BC in early and advanced settings to ascertain which populations will most benefit from these drugs. Of the biomarkers that were analysed, such as PIK3CA, AKT, PTEN mutations, insulin levels, 18 F-FDG-PET/TC, only the PIK3CA-mutations (PIK3CA-mut) and the AKT pathway alterations seem to have a predictive value for treatments with alpelisib and capivasertib. However, due to the retrospective and exploratory nature of the study, the data did not provide conclusive results. In addition, the different methods used to detect PIK3CA/AKT1/PTEN alterations underline the fact that the optimal diagnostic companion has yet to be established. We have summarised the clinical data on the approved and discontinued agents targeting this pathway and have assessed the drugs development, successes, and failures. Finally, because of tumour heterogeneity, we emphasise the importance of reassessing the mutational status of PI3KCA in both metastatic tissue and blood at the time of disease progression to better tailor treatment for patients.
Collapse
Affiliation(s)
- M Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - F Jacobs
- Humanitas Clinical and Research Center - IRCCS, Humanitas Cancer Center, via Manzoni 56, 20089 Rozzano, Milan, Italy; Early Phase Trials Unit Institut Bergonié Bordeaux, France
| | - C Molinelli
- Early Phase Trials Unit Institut Bergonié Bordeaux, France; Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy; Department of Medical Oncology, U.O. Clinical di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - V Debien
- Early Phase Trials Unit Institut Bergonié Bordeaux, France
| | - H Faith Dewhurst
- Faculty of Medicine, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - M Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - F Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - C Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - U De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | | |
Collapse
|
4
|
Chen R, Yu Y, Zhang J, Song C, Wang C. Efficacy and safety of neoadjuvant therapy for HR-positive/HER2-negative early breast cancer: a Bayesian network meta-analysis. Expert Rev Anticancer Ther 2024; 24:599-611. [PMID: 38693054 DOI: 10.1080/14737140.2024.2350105] [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: 12/20/2023] [Accepted: 04/25/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Neoadjuvant treatment for hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) breast cancer is controversial and requires a comprehensive analysis for optimal therapy assessment. Therefore, a two-step Bayesian network meta-analysis (NMA) was performed to compare the efficacy and safety of different neoadjuvant regimens. RESEARCH DESIGN AND METHODS Phase II/III randomized clinical trials comparing various neoadjuvant therapies for HR+/HER2- breast cancer were included. NMA and pairwise meta-analyses were conducted using Stata (version 14), R (version 4.2.3), and Review Manager 5.4. RESULTS Twenty-eight studies (5,625 patients) were eligible. NMA of objective response rate (ORR) indicated the highest SUCRA for chemotherapy (CT) and chemotherapy with anthracycline (CT(A)). Pathologic complete response (PCR) NMA demonstrated significant PCR improvement with chemotherapy regimens containing programmed cell death protein-1 and programmed cell death ligand-1 inhibitors (PD-1i/PD-L1i) and poly ADP-ribose polymerase inhibitors (PARPi). Combined analysis considering both the ORR and safety highlighted CT(A)'s efficacy and toxicity balance. CONCLUSIONS CT(A) and CT showed improved ORR compared with alternative regimens. CT(A) combined with PD-1/PD-L1 or PARP inhibitors significantly increased PCR rates. Comprehensive assessment of both ORR and safety indicated that CT(A) represents an optimal neoadjuvant therapy for HR+/HER2- breast cancer, whereas AI + CDK4/6 inhibitors rank solely behind chemotherapy. REGISTRATION PROSPERO Registration: CRD42024538948. International Platform of Registered Systematic Review and Meta-Analysis Protocols (INPLASY) registration number INPLASY202440092.
Collapse
Affiliation(s)
- Ruiliang Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yushuai Yu
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chuangui Song
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
5
|
Van Cauwenberge J, Van Baelen K, Maetens M, Geukens T, Nguyen HL, Nevelsteen I, Smeets A, Deblander A, Neven P, Koolen S, Wildiers H, Punie K, Desmedt C. Reporting on patient's body mass index (BMI) in recent clinical trials for patients with breast cancer: a systematic review. Breast Cancer Res 2024; 26:81. [PMID: 38778365 PMCID: PMC11112918 DOI: 10.1186/s13058-024-01832-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The proportion of patients with breast cancer and obesity is increasing. While the therapeutic landscape of breast cancer has been expanding, we lack knowledge about the potential differential efficacy of most drugs according to the body mass index (BMI). Here, we conducted a systematic review on recent clinical drug trials to document the dosing regimen of recent drugs, the reporting of BMI and the possible exclusion of patients according to BMI, other adiposity measurements and/or diabetes (leading comorbidity of obesity). We further explored whether treatment efficacy was evaluated according to BMI. METHODS A search of Pubmed and ClinicalTrials.gov was performed to identify phase I-IV trials investigating novel systemic breast cancer treatments. Dosing regimens and exclusion based on BMI, adiposity measurements or diabetes, documentation of BMI and subgroup analyses according to BMI were assessed. RESULTS 495 trials evaluating 26 different drugs were included. Most of the drugs (21/26, 81%) were given in a fixed dose independent of patient weight. BMI was an exclusion criterion in 3 out of 495 trials. Patients with diabetes, the leading comorbidity of obesity, were excluded in 67/495 trials (13.5%). Distribution of patients according to BMI was mentioned in 8% of the manuscripts, subgroup analysis was performed in 2 trials. No other measures of adiposity/body composition were mentioned in any of the trials. Retrospective analyses on the impact of BMI were performed in 6 trials. CONCLUSIONS Patient adiposity is hardly considered as most novel drug treatments are given in a fixed dose. BMI is generally not reported in recent trials and few secondary analyses are performed. Given the prevalence of patients with obesity and the impact obesity can have on pharmacokinetics and cancer biology, more attention should be given by investigators and study sponsors to reporting patient's BMI and evaluating its impact on treatment efficacy and toxicity.
Collapse
Affiliation(s)
- Josephine Van Cauwenberge
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Karen Van Baelen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Ha Linh Nguyen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
| | - Ines Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Anne Deblander
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Stijn Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Kevin Punie
- Department of Medical Oncology, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium.
| |
Collapse
|
6
|
Wu J, Wen T, Marzio A, Song D, Chen S, Yang C, Zhao F, Zhang B, Zhao G, Ferri A, Cheng H, Ma J, Ren H, Chen QY, Yang Y, Qin S. FBXO32-mediated degradation of PTEN promotes lung adenocarcinoma progression. Cell Death Dis 2024; 15:282. [PMID: 38643215 PMCID: PMC11032391 DOI: 10.1038/s41419-024-06635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 03/21/2024] [Accepted: 03/28/2024] [Indexed: 04/22/2024]
Abstract
FBXO32, a member of the F-box protein family, is known to play both oncogenic and tumor-suppressive roles in different cancers. However, the functions and the molecular mechanisms regulated by FBXO32 in lung adenocarcinoma (LUAD) remain unclear. Here, we report that FBXO32 is overexpressed in LUAD compared with normal lung tissues, and high expression of FBXO32 correlates with poor prognosis in LUAD patients. Firstly, we observed with a series of functional experiments that FBXO32 alters the cell cycle and promotes the invasion and metastasis of LUAD cells. We further corroborate our findings using in vivo mouse models of metastasis and confirmed that FBXO32 positively regulates LUAD tumor metastasis. Using a proteomic-based approach combined with computational analyses, we found a positive correlation between FBXO32 and the PI3K/AKT/mTOR pathway, and identified PTEN as a FBXO32 interactor. More important, FBXO32 binds PTEN via its C-terminal substrate binding domain and we also validated PTEN as a bona fide FBXO32 substrate. Finally, we demonstrated that FBXO32 promotes EMT and regulates the cell cycle by targeting PTEN for proteasomal-dependent degradation. In summary, our study highlights the role of FBXO32 in promoting the PI3K/AKT/mTOR pathway via PTEN degradation, thereby fostering lung adenocarcinoma progression.
Collapse
Affiliation(s)
- Jie Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Ting Wen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Antonio Marzio
- Department of Pathology and Laboratory Medicine, Meyer Cancer Center, Weill Cornell Medical Center, New York, NY, USA
| | - Dingli Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Sisi Chen
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chengcheng Yang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengyu Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Boxiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guang Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Alessandra Ferri
- Department of Pathology and Laboratory Medicine, Meyer Cancer Center, Weill Cornell Medical Center, New York, NY, USA
| | - Hao Cheng
- Department of Rehabilitation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiao Ma
- Department of Rehabilitation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hong Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiao Yi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yiping Yang
- Clinical Research Center for Shaanxi Provincial Radiotherapy, Department of Radiation Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi, China.
| | - Sida Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Biobank, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Browne IM, André F, Chandarlapaty S, Carey LA, Turner NC. Optimal targeting of PI3K-AKT and mTOR in advanced oestrogen receptor-positive breast cancer. Lancet Oncol 2024; 25:e139-e151. [PMID: 38547898 DOI: 10.1016/s1470-2045(23)00676-9] [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: 09/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 04/02/2024]
Abstract
The growing availability of targeted therapies for patients with advanced oestrogen receptor-positive breast cancer has improved survival, but there remains much to learn about the optimal management of these patients. The PI3K-AKT and mTOR pathways are among the most commonly activated pathways in breast cancer, whose crucial role in the pathogenesis of this tumour type has spurred major efforts to target this pathway at specific kinase hubs. Approvals for oestrogen receptor-positive advanced breast cancer include the PI3K inhibitor alpelisib for PIK3CA-mutated tumours, the AKT inhibitor capivasertib for tumours with alterations in PIK3CA, AKT1, or PTEN, and the mTOR inhibitor everolimus, which is used irrespective of mutation status. The availability of different inhibitors leaves physicians with a potentially challenging decision over which of these therapies should be used for individual patients and when. In this Review, we present a comprehensive summary of our current understanding of the pathways and the three inhibitors and discuss strategies for the optimal sequencing of therapies in the clinic, particularly after progression on a CDK4/6 inhibitor.
Collapse
Affiliation(s)
- Iseult M Browne
- Breast Cancer Now Research Centre, Institute of Cancer Research, London, UK; Ralph Lauren Centre for Breast Cancer Research and Breast Unit, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Fabrice André
- Department of Medical Oncology, INSERM U981, Institut Gustave Roussy, Université Paris Saclay, Villejuif, France
| | | | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Nicholas C Turner
- Breast Cancer Now Research Centre, Institute of Cancer Research, London, UK; Ralph Lauren Centre for Breast Cancer Research and Breast Unit, The Royal Marsden Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
8
|
Ketcham JM, Harwood SJ, Aranda R, Aloiau AN, Bobek BM, Briere DM, Burns AC, Caddell Haatveit K, Calinisan A, Clarine J, Elliott A, Engstrom LD, Gunn RJ, Ivetac A, Jones B, Kuehler J, Lawson JD, Nguyen N, Parker C, Pearson KE, Rahbaek L, Saechao B, Wang X, Waters A, Waters L, Watkins AH, Olson P, Smith CR, Christensen JG, Marx MA. Discovery of Pyridopyrimidinones that Selectively Inhibit the H1047R PI3Kα Mutant Protein. J Med Chem 2024; 67:4936-4949. [PMID: 38477582 PMCID: PMC10983000 DOI: 10.1021/acs.jmedchem.4c00078] [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: 01/10/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
The H1047R mutation of PIK3CA is highly prevalent in breast cancers and other solid tumors. Selectively targeting PI3KαH1047R over PI3KαWT is crucial due to the role that PI3KαWT plays in normal cellular processes, including glucose homeostasis. Currently, only one PI3KαH1047R-selective inhibitor has progressed into clinical trials, while three pan mutant (H1047R, H1047L, H1047Y, E542K, and E545K) selective PI3Kα inhibitors have also reached the clinical stage. Herein, we report the design and discovery of a series of pyridopyrimidinones that inhibit PI3KαH1047R with high selectivity over PI3KαWT, resulting in the discovery of compound 17. When dosed in the HCC1954 tumor model in mice, 17 provided tumor regressions and a clear pharmacodynamic response. X-ray cocrystal structures from several PI3Kα inhibitors were obtained, revealing three distinct binding modes within PI3KαH1047R including a previously reported cryptic pocket in the C-terminus of the kinase domain wherein we observe a ligand-induced interaction with Arg1047.
Collapse
Affiliation(s)
| | | | - Ruth Aranda
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Athenea N. Aloiau
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Briana M. Bobek
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - David M. Briere
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Aaron C. Burns
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | | | - Andrew Calinisan
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Jeffery Clarine
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Adam Elliott
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Lars D. Engstrom
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Robin J. Gunn
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Anthony Ivetac
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Benjamin Jones
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Jon Kuehler
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - J. David Lawson
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Natalie Nguyen
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Cody Parker
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Kelly E. Pearson
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Lisa Rahbaek
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Barbara Saechao
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Xiaolun Wang
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Anna Waters
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Laura Waters
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Ashlee H. Watkins
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Peter Olson
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Christopher R. Smith
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - James G. Christensen
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| | - Matthew A. Marx
- Mirati Therapeutics, 3545 Cray Court, San Diego, California 92121, United States
| |
Collapse
|
9
|
Shan KS, Bonano-Rios A, Theik NWY, Hussein A, Blaya M. Molecular Targeting of the Phosphoinositide-3-Protein Kinase (PI3K) Pathway across Various Cancers. Int J Mol Sci 2024; 25:1973. [PMID: 38396649 PMCID: PMC10888452 DOI: 10.3390/ijms25041973] [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: 01/01/2024] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of the phosphatidylinositol-3-kinase (PI3K) pathway can lead to uncontrolled cellular growth and tumorigenesis. Targeting PI3K and its downstream substrates has been shown to be effective in preclinical studies and phase III trials with the approval of several PI3K pathway inhibitors by the Food and Drug Administration (FDA) over the past decade. However, the limited clinical efficacy of these inhibitors, intolerable toxicities, and acquired resistances limit the clinical application of PI3K inhibitors. This review discusses the PI3K signaling pathway, alterations in the PI3K pathway causing carcinogenesis, current and novel PI3K pathway inhibitors, adverse effects, resistance mechanisms, challenging issues, and future directions of PI3K pathway inhibitors.
Collapse
Affiliation(s)
- Khine S. Shan
- Division of Hematology and Oncology, Memorial Health Care, Pembroke Pines, FL 33028, USA; (A.B.-R.); (A.H.); (M.B.)
| | - Amalia Bonano-Rios
- Division of Hematology and Oncology, Memorial Health Care, Pembroke Pines, FL 33028, USA; (A.B.-R.); (A.H.); (M.B.)
| | - Nyein Wint Yee Theik
- Division of Internal Medicine, Memorial Health Care, Pembroke Pines, FL 33028, USA;
| | - Atif Hussein
- Division of Hematology and Oncology, Memorial Health Care, Pembroke Pines, FL 33028, USA; (A.B.-R.); (A.H.); (M.B.)
| | - Marcelo Blaya
- Division of Hematology and Oncology, Memorial Health Care, Pembroke Pines, FL 33028, USA; (A.B.-R.); (A.H.); (M.B.)
| |
Collapse
|
10
|
Cantini L, Trapani D, Guidi L, Boscolo Bielo L, Scafetta R, Koziej M, Vidal L, Saini KS, Curigliano G. Neoadjuvant therapy in hormone Receptor-Positive/HER2-Negative breast cancer. Cancer Treat Rev 2024; 123:102669. [PMID: 38141462 DOI: 10.1016/j.ctrv.2023.102669] [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: 09/20/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/25/2023]
Abstract
Neoadjuvant therapy is commonly used in patients with locally advanced or inoperable breast cancer (BC). Neoadjuvant chemotherapy (NACT) represents an established treatment modality able to downstage tumours, facilitate breast-conserving surgery, yet also achieve considerable pathologic complete response (pCR) rates in HER2-positive and triple-negative BC. For patients with HR+/HER2- BC, the choice between NACT and neoadjuvant endocrine therapy (NET) is still based on clinical and pathological features and not guided by biomarkers of defined clinical utility, differently from the adjuvant setting where gene-expression signatures have been widely adopted to drive decision-making. In this review, we summarize the evidence supporting the choice of NACT vs NET in HR+/HER2- BC, discussing the issues surrounding clinical trial design and proper selection of patients for every treatment. It is time to question the binary paradigm of responder vs non-responders as well as the "one size fits all" approach in luminal BC, supporting the utilization of continuous endpoints and the adoption of tissue and plasma-based biomarkers at multiple timepoints. This will eventually unleash the full potential of neoadjuvant therapy which is to modulate patient treatment based on treatment sensitivity and surgical outcomes. We also reviewed the current landscape of neoadjuvant studies for HR+/HER2- BC, focusing on antibody-drug conjugates (ADCs) and immunotherapy combinations. Finally, we proposed a roadmap for future neoadjuvant approaches in HR+/HER2- BC, which should be based on a staggered biomarker-driven treatment selection aiming at impacting long-term relevant endpoints.
Collapse
Affiliation(s)
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenzo Guidi
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Luca Boscolo Bielo
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Roberta Scafetta
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; Department of medical oncology, Campus Bio-Medico, University of Rome, Rome, Italy
| | | | | | | | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of New Drugs and Early Drug Development, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy.
| |
Collapse
|
11
|
Wang Y, Du X, Xin H, Xu R. Efficacy and Safety of Phosphatidylinositol 3-kinase Inhibitors for Patients with Breast Cancer: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Curr Cancer Drug Targets 2024; 24:941-951. [PMID: 38275057 DOI: 10.2174/0115680096266181231207110048] [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: 08/05/2023] [Revised: 10/16/2023] [Accepted: 11/13/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Phosphatidylinositol 3-kinase (PI3K) inhibitors belong to the class of drugs that inhibit the activity of the PI3K protein, which is commonly overexpressed in breast cancer cells. However, there is a need to summarize the evidence to provide conclusive advice on the benefit of PI3K inhibitors in breast cancer patients. Therefore, this review assessed the effectiveness and safety of the PI3K inhibitors amongst breast cancer patients. METHODS Searches were made in PubMed Central, EMBASE, MEDLINE, SCOPUS, CENTRAL, WHO trial registry and Clinicaltrials.gov up to December 2022. Meta-analysis was executed using the random-effects model. Pooled hazard ratio (HR)/risk ratio (RR) was reported with 95% confidence intervals (CIs). RESULTS In total, 13 studies were included in the analysis. Most were multi-country studies and had a higher risk of bias. Regarding the efficacy parameters, pooled HR for progression-free survival was 0.79 (95%CI: 0.67-0.92), pooled RR for complete response was 1.54 [95%CI: 1.14 to 2.09], partial response was 1.18 [95%CI: 0.87-1.61], overall response was 1.20 [95%CI: 0.93-1.56], stable disease was 1.09 [95%CI: 0.78-1.53], progressive disease was 0.80 [95%CI: 0.74 to 0.87], and clinical benefit was 1.08 [95%CI: 0.80-1.49]. For safety parameters, pooled RR for hyperglycemia was 4.57 [95%CI: 3.15-6.62], and gastrointestinal toxicity was 1.82 [95%CI: 1.56 to 2.14]. CONCLUSION PI3K inhibitors had better efficacy than the present standard of concern for patients with breast cancer, especially among patients with PIK3CA mutations. Hence, clinicians and oncologists can provide this drug for the target population with extra caution for diabetes patients.
Collapse
Affiliation(s)
- Yi Wang
- Department of Nail and Breast Surgery, Panzhihua Hospital of Integrated Traditional Chinese and Western Medicine (Affiliated Hospital of Panzhihua University, Panzhihua City, Sichuan Province, 617000, China
| | - Xianling Du
- Department of Oncology, The Central Hospital of Enshi Tujia Ang Miao Autonomous Prefecture (Enshi Clinical College of Wuhan University), Enshi City,Hubei Province, 445000, China
| | - Hongqiang Xin
- Department of two gland surgery, Qilu Hospital Huantai Branch, People's Hospital of Huantai County, Zibo city, Shandong Province, 256400, China
| | - Ruimin Xu
- Department of Anorectal, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, Shanghai City, 212000, China
| |
Collapse
|
12
|
Jackisch C, Argyriadis A. Endocrine Therapy: From Ovarian Ablation to Individualized Therapy and Signal Inhibition. Breast Care (Basel) 2023; 18:440-447. [PMID: 38125923 PMCID: PMC10730102 DOI: 10.1159/000534154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/13/2023] [Indexed: 12/23/2023] Open
Abstract
Background At the end of the 19th century, Sir George Thomas Beatson first discovered the positive influence of a bilateral oophorectomy on the development of breast cancer lesions in women with advanced disease. Since then, endocrine therapy has been a key component of the treatment of both early (EBC) and advanced-stage (MBC) hormone receptor (HR)-positive breast cancer. Summary This review discusses the evolution of this therapeutic approach from the introduction of high-dose estrogen therapy leading to the development of several antiestrogen therapies. Recently, the new generation of drugs includes selective estrogen receptor modulators, orally administered selective estrogen receptor degraders (SERDs), as well as more unique agents such as complete estrogen receptor antagonists, proteolysis targeting chimeric, and selective estrogen receptor covalent antagonists. These drugs are under evaluation in various levels of randomized clinical trials (RCTs) being evaluated in both early and metastatic settings. As of today, the options in EBC are ranging from short-term neoadjuvant endocrine therapy to monitor the responsiveness of Ki-67 to combined endocrine therapy in MBC, introducing the combination of endocrine therapy and CDK4/6 inhibition as well as PARP inhibition in patients with luminal breast cancer presenting with germline BRCA1/2 mutations. The results of global RCTs are settled in global and local guidelines to optimize the individual therapy of our patients with luminal EBC. Key Messages Endocrine intervention in hormone-sensitive breast cancer remains one of the most important options in all settings of early and metastatic breast cancer.
Collapse
Affiliation(s)
- Christian Jackisch
- Department of Obstetrics and Gynaecology, Sana Klinikum Offenbach, Offenbach, Germany
| | - Athanasios Argyriadis
- Department of Obstetrics and Gynaecology, Sana Klinikum Offenbach, Offenbach, Germany
| |
Collapse
|
13
|
Jia W, Luo S, Guo H, Kong D. Development of PI3Kα inhibitors for tumor therapy. J Biomol Struct Dyn 2023; 41:8587-8604. [PMID: 36221910 DOI: 10.1080/07391102.2022.2132293] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/28/2022] [Indexed: 10/17/2022]
Abstract
The PI3K/AKT/mTOR signaling pathway is well known to be involved in cell growth, proliferation, metabolism and other cellular physiological processes. Abnormal activation of this pathway is closely related to tumorigenesis and metastasis. As the starting node of the pathway, PI3K is known to contain 4 isoforms, including PI3Kα, a heterodimer composed of the catalytic subunit p110α and the regulatory subunit p85. PIK3CA, which encodes p110α, is frequently mutated in cancer, especially breast cancer. Abnormal activation of PI3Kα promotes cancer cell proliferation, migration, invasion, and angiogenesis; therefore, PI3Kα has become a key target for the development of anticancer drugs. The hinge region and the region of the mutation site in the PI3Kα protein are important for designing PI3Kα-specific inhibitors. As the group shared by the most PI3Kα-specific inhibitors reported thus far, carboxamide can produce hydrogen bonds with Gln859 and Ser854. Gln859 is specific to the p110α protein in producing hydrogen bond interactions with PI3Kα-specific inhibitors and this is a key point for designing PI3Kα inhibitors. To date, alpelisib is the only PI3Kα inhibitor approved for the treatment of breast cancer. Several other PI3Kα inhibitors are under evaluation in clinical trials. In this review, we briefly describe PI3Kα and its role in tumorigenesis, summarize the clinical trial results of some PI3Kα inhibitors as well as the synthetic routes of alpelisib, and finally give our proposal for the development of novel PI3Kα inhibitors for tumor therapy. HighlightsWe summarize the progress of PI3Kα and PI3Kα inhibitors in cancer from the second half of the 20th century to the present.We describe the clinical trial results of PI3Kα inhibitors as well as the synthetic routes of the only approved PI3Kα inhibitor alpelisib.Crystal structure of alpelisib bound to the PI3Kα receptor binding domain.This review gives proposal for the development of novel PI3Kα inhibitors and will serve as a complementary summary to other reviews in the research field of PI3K inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wenqing Jia
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shuyu Luo
- School of Stomatology, Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Han Guo
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Jeong H, Kim SB. Neoadjuvant endocrine therapy in ER-positive breast cancer: evolution, indication, and tailored treatment strategy. Ther Adv Med Oncol 2023; 15:17588359231200457. [PMID: 37786536 PMCID: PMC10541763 DOI: 10.1177/17588359231200457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 08/25/2023] [Indexed: 10/04/2023] Open
Abstract
In recent years, endocrine therapy (ET), an effective systemic treatment for the management of estrogen receptor (ER)-positive breast cancers, has regained interest as a neoadjuvant therapy based on evidence that ET can fulfill the aim of neoadjuvant systemic treatment for tumor shrinkage as well as elucidate important clinical information on endocrine sensitivity that enables the prognostication of patients. Moreover, neoadjuvant endocrine therapy (NET) potentially provides an opportunity for early assessment of the clinical efficacy of novel agents. Furthermore, recently reported trials have generated evidence for a more tailored approach for perioperative management of ER-positive breast cancer using clinical and molecular biomarkers, and this has provided a rationale that enables the broadening of clinical indications for NET. This review discusses the current evidence for NET, the evolution of NET trials, clinical indications, and NET-based treatment strategies.
Collapse
Affiliation(s)
- Hyehyun Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 138-736, Republic of Korea
| |
Collapse
|
15
|
Glaviano A, Foo ASC, Lam HY, Yap KCH, Jacot W, Jones RH, Eng H, Nair MG, Makvandi P, Geoerger B, Kulke MH, Baird RD, Prabhu JS, Carbone D, Pecoraro C, Teh DBL, Sethi G, Cavalieri V, Lin KH, Javidi-Sharifi NR, Toska E, Davids MS, Brown JR, Diana P, Stebbing J, Fruman DA, Kumar AP. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer 2023; 22:138. [PMID: 37596643 PMCID: PMC10436543 DOI: 10.1186/s12943-023-01827-6] [Citation(s) in RCA: 275] [Impact Index Per Article: 275.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
The PI3K/AKT/mTOR (PAM) signaling pathway is a highly conserved signal transduction network in eukaryotic cells that promotes cell survival, cell growth, and cell cycle progression. Growth factor signalling to transcription factors in the PAM axis is highly regulated by multiple cross-interactions with several other signaling pathways, and dysregulation of signal transduction can predispose to cancer development. The PAM axis is the most frequently activated signaling pathway in human cancer and is often implicated in resistance to anticancer therapies. Dysfunction of components of this pathway such as hyperactivity of PI3K, loss of function of PTEN, and gain-of-function of AKT, are notorious drivers of treatment resistance and disease progression in cancer. In this review we highlight the major dysregulations in the PAM signaling pathway in cancer, and discuss the results of PI3K, AKT and mTOR inhibitors as monotherapy and in co-administation with other antineoplastic agents in clinical trials as a strategy for overcoming treatment resistance. Finally, the major mechanisms of resistance to PAM signaling targeted therapies, including PAM signaling in immunology and immunotherapies are also discussed.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Aaron S C Foo
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore
| | - Hiu Y Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Kenneth C H Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - William Jacot
- Department of Medical Oncology, Institut du Cancer de Montpellier, Inserm U1194, Montpellier University, Montpellier, France
| | - Robert H Jones
- Cardiff University and Velindre Cancer Centre, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Huiyan Eng
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bangalore, 560034, India
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Inserm U1015, Université Paris-Saclay, Paris, France
| | - Matthew H Kulke
- Section of Hematology and Medical Oncology, Boston University and Boston Medical Center, Boston, MA, USA
| | - Richard D Baird
- Cancer Research UK Cambridge Centre, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bangalore, 560034, India
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Camilla Pecoraro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Daniel B L Teh
- Departments of Ophthalmology and Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, and Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kevin H Lin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Eneda Toska
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Justin Stebbing
- Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, 216 Sprague Hall, Irvine, CA, USA
| | - Alan P Kumar
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
16
|
Schutte T, Zeverijn LJ, Geurts BS, de Wit GF, Kok M, Opdam FL. Beyond Skin Rash: Alpelisib-Induced Anaphylactic Reactions. Oncologist 2023:7136676. [PMID: 37086483 DOI: 10.1093/oncolo/oyad092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/09/2023] [Indexed: 04/24/2023] Open
Abstract
Alpelisib is a specific oral PI3K inhibitor used combined with fulvestrant for the treatment of patients with HR+/HER2-/PIK3CA-mutated metastatic breast cancer. Adverse drug reactions with alpelisib are common, including hyperglycemia and rash. Here we describe extraordinary and life-threatening reactions beyond skin rash in two patients with progressive PIK3CA-mutated metastatic cancer in whom alpelisib was initiated. Case-A (vaginal cancer): After 10 days on treatment, she developed dry eyes, generalized rash and itching. Alpelisib was interrupted and symptomatic treatment initiated. Because of an initial tumor response, a rechallenge was done. Ninety minutes after a reduced dose of alpelisib, she developed an anaphylactic reaction with angioedema, hypotension, and skin rash. Case-B (breast cancer): After 11 days on treatment, she developed skin rash and alpelisib was interrupted. At re-initiation, she felt tingles in her face and ears and some skin erythema. Given the mild rash, a second rechallenge with premedication was performed. Ninety minutes after a reduced dose of alpelisib, she developed a type-1 allergic reaction with angioedema, tingles, and skin rash. In both cases, a type-1 allergic reaction was diagnosed and symptomatic treatment was initiated, alpelisib was permanently discontinued and the patients fully recovered the next week(s). This report underlines the critical importance to consider type-I allergic reactions in the differential diagnosis in cases of rash associated with alpelisib. Even if a reaction develops after days on treatment, a type-I allergic reaction cannot be excluded. A rechallenge can be dangerous and should always be well contemplated or even avoided.
Collapse
Affiliation(s)
- Tim Schutte
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Medical Oncology, de Boelelaan, Amsterdam, The Netherlands
| | - Laurien J Zeverijn
- The Netherlands Cancer Institute, Division of Molecular Oncology and Immunology, CX Amsterdam, The Netherlands
| | - Birgit S Geurts
- The Netherlands Cancer Institute, Division of Molecular Oncology and Immunology, CX Amsterdam, The Netherlands
| | - Gijsbrecht F de Wit
- The Netherlands Cancer Institute, Division of Molecular Oncology and Immunology, CX Amsterdam, The Netherlands
| | - Marleen Kok
- The Netherlands Cancer Institute, Department of Medical Oncology, CX Amsterdam, The Netherlands
| | - Frans L Opdam
- The Netherlands Cancer Institute, Department of Medical Oncology, CX Amsterdam, The Netherlands
| |
Collapse
|
17
|
Viganò M, La Milia M, Grassini MV, Pugliese N, De Giorgio M, Fagiuoli S. Hepatotoxicity of Small Molecule Protein Kinase Inhibitors for Cancer. Cancers (Basel) 2023; 15:cancers15061766. [PMID: 36980652 PMCID: PMC10046041 DOI: 10.3390/cancers15061766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Small molecule protein kinase inhibitors (PKIs) have become an effective strategy for cancer patients. However, hepatotoxicity is a major safety concern of these drugs, since the majority are reported to increase transaminases, and few of them (Idelalisib, Lapatinib, Pazopanib, Pexidartinib, Ponatinib, Regorafenib, Sunitinib) have a boxed label warning. The exact rate of PKI-induced hepatoxicity is not well defined due to the fact that the majority of data arise from pre-registration or registration trials on fairly selected patients, and the post-marketing data are often based only on the most severe described cases, whereas most real practice studies do not include drug-related hepatotoxicity as an end point. Although these side effects are usually reversible by dose adjustment or therapy suspension, or by switching to an alternative PKI, and fatality is uncommon, all patients undergoing PKIs should be carefully pre-evaluated and monitored. The management of this complication requires an individually tailored reappraisal of the risk/benefit ratio, especially in patients who are responding to therapy. This review reports the currently available data on the risk and management of hepatotoxicity of all the approved PKIs.
Collapse
Affiliation(s)
- Mauro Viganò
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Correspondence: ; Tel.: +39-035-2674259; Fax: +39-035-2674964
| | - Marta La Milia
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Maria Vittoria Grassini
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Section of Gastroenterology & Hepatology, Department of Health Promotion Sciences Maternal and Infant Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, 90127 Palermo, Italy
| | - Nicola Pugliese
- Department of Gastroenterology, Division of Internal Medicine and Hepatology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Massimo De Giorgio
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Stefano Fagiuoli
- Gastroenterology Hepatology and Transplantation Unit, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
- Gastroenterology, Department of Medicine, University of Milan Bicocca, 20126 Milan, Italy
| |
Collapse
|
18
|
Alves CL, Ditzel HJ. Drugging the PI3K/AKT/mTOR Pathway in ER+ Breast Cancer. Int J Mol Sci 2023; 24:4522. [PMID: 36901954 PMCID: PMC10003259 DOI: 10.3390/ijms24054522] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
The frequent activation of the PI3K/AKT/mTOR pathway and its crucial role in estrogen receptor-positive (ER+) breast cancer tumorigenesis and drug resistance has made it a highly attractive therapeutic target in this breast cancer subtype. Consequently, the number of new inhibitors in clinical development targeting this pathway has drastically increased. Among these, the PIK3CA isoform-specific inhibitor alpelisib and the pan-AKT inhibitor capivasertib were recently approved in combination with the estrogen receptor degrader fulvestrant for the treatment of ER+ advanced breast cancer after progression on an aromatase inhibitor. Nevertheless, the clinical development of multiple inhibitors of the PI3K/AKT/mTOR pathway, in parallel with the incorporation of CDK4/6 inhibitors into the standard of care treatment in ER+ advanced breast cancer, has led to a multitude of available therapeutic agents and many possible combined strategies which complicate personalizing treatment. Here, we review the role of the PI3K/AKT/mTOR pathway in ER+ advanced breast cancer, highlighting the genomic contexts in which the various inhibitors of this pathway may have superior activity. We also discuss selected trials with agents targeting the PI3K/AKT/mTOR and related pathways as well as the rationale supporting the clinical development of triple combination therapy targeting ER, CDK4/6 and PI3K/AKT/mTOR in ER+ advanced breast cancer.
Collapse
Affiliation(s)
- Carla L. Alves
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Henrik J. Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Oncology, Institute of Clinical Research, Odense University Hospital, 5000 Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
19
|
Bertucci A, Bertucci F, Gonçalves A. Phosphoinositide 3-Kinase (PI3K) Inhibitors and Breast Cancer: An Overview of Current Achievements. Cancers (Basel) 2023; 15:1416. [PMID: 36900211 PMCID: PMC10001361 DOI: 10.3390/cancers15051416] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 02/26/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) pathway is one of the most altered pathways in human cancers, and it plays a central role in cellular growth, survival, metabolism, and cellular mobility, making it a particularly interesting therapeutic target. Recently, pan-inhibitors and then selective p110α subunit inhibitors of PI3K were developed. Breast cancer is the most frequent cancer in women and, despite therapeutic progress in recent years, advanced breast cancers remain incurable and early breast cancers are at risk of relapse. Breast cancer is divided in three molecular subtypes, each with its own molecular biology. However, PI3K mutations are found in all breast cancer subtypes in three main "hotspots". In this review, we report the results of the most recent and main ongoing studies evaluating pan-PI3K inhibitors and selective PI3K inhibitors in each breast cancer subtype. In addition, we discuss the future of their development, the various potential mechanisms of resistance to these inhibitors and the ways to circumvent them.
Collapse
Affiliation(s)
| | | | - Anthony Gonçalves
- Medical Oncology Department, CRCM, INSERM, CNRS, Institut Paoli-Calmettes, Aix-Marseille University, 13009 Marseille, France
| |
Collapse
|
20
|
Sirico M, Virga A, Conte B, Urbini M, Ulivi P, Gianni C, Merloni F, Palleschi M, Gasperoni M, Curcio A, Saha D, Buono G, Muñoz M, De Giorgi U, Schettini F. Neoadjuvant endocrine therapy for luminal breast tumors: State of the art, challenges and future perspectives. Crit Rev Oncol Hematol 2023; 181:103900. [PMID: 36565894 DOI: 10.1016/j.critrevonc.2022.103900] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Neoadjuvant endocrine treatment (NET) associates to satisfactory rates of breast conservative surgery and conversions from inoperable to operable hormone receptor-positive (HR+)/HER2-negative breast cancer (BC), with less toxicities than neoadjuvant chemotherapy (NACT) and similar outcomes. Hence, it has been proposed as a logical alternative to NACT in patients with HR+/HER2- BC candidate to a neoadjuvant approach. Nevertheless, potential barriers to the widespread use of NET include the heterogeneous nature of patient response coupled with the long duration needed to achieve a clinical response. However, interest in NET has significantly increased in the last decade, owing to more in-depth investigation of several biomarkers for a more adequate patient selection and on-treatment benefit monitoring, such as PEPI score, Ki67 and genomic assays. This review is intended to describe the state-of-the-art regarding NET, its future perspectives and potential integration with molecular biomarkers for the optimal selection of patients, regimen and duration of (neo)adjuvant treatments.
Collapse
Affiliation(s)
- Marianna Sirico
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandra Virga
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Benedetta Conte
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Filippo Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Marco Gasperoni
- Breast Surgery Unit, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Annalisa Curcio
- Breast Surgery Unit, Morgagni-Pierantoni Hospital, AUSL Romagna, Forlì, Italy
| | - Debjani Saha
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Giuseppe Buono
- Department of Breast and Thoracic Oncology, National Cancer Institute, Fondazione G. Pascale, Naples, Italy
| | - Montserrat Muñoz
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain; Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesco Schettini
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain; Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain; Faculty of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Martínez-Pérez C, Turnbull AK, Kay C, Dixon JM. Neoadjuvant endocrine therapy in postmenopausal women with HR+/HER2- breast cancer. Expert Rev Anticancer Ther 2023; 23:67-86. [PMID: 36633402 DOI: 10.1080/14737140.2023.2162043] [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: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
INTRODUCTION While endocrine therapy is the standard-of-care adjuvant treatment for hormone receptor-positive (HR+) breast cancers, there is also extensive evidence for the role of pre-operative (or neoadjuvant) endocrine therapy (NET) in HR+ postmenopausal women. AREAS COVERED We conducted a thorough review of the published literature, to summarize the evidence to date, including studies of how NET compares to neoadjuvant chemotherapy, which NET agents are preferable, and the optimal duration of NET. We describe the importance of on-treatment assessment of response, the different predictors available (including Ki67, PEPI score, and molecular signatures) and the research opportunities the pre-operative setting offers. We also summarize recent combination trials and discuss how the COVID-19 pandemic led to increases in NET use for safe management of cases with deferred surgery and adjuvant treatments. EXPERT OPINION NET represents a safe and effective tool for the management of postmenopausal women with HR+/HER2- breast cancer, enabling disease downstaging and a wider range of surgical options. Aromatase inhibitors are the preferred NET, with evidence suggesting that longer regimens might yield optimal results. However, NET remains currently underutilised in many territories and institutions. Further validation of predictors for treatment response and benefit is needed to help standardise and fully exploit the potential of NET in the clinic.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Arran K Turnbull
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Charlene Kay
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - J Michael Dixon
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, Scotland
| |
Collapse
|
22
|
Cerma K, Piacentini F, Moscetti L, Barbolini M, Canino F, Tornincasa A, Caggia F, Cerri S, Molinaro A, Dominici M, Omarini C. Targeting PI3K/AKT/mTOR Pathway in Breast Cancer: From Biology to Clinical Challenges. Biomedicines 2023; 11:biomedicines11010109. [PMID: 36672617 PMCID: PMC9855880 DOI: 10.3390/biomedicines11010109] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023] Open
Abstract
Breast cancer (BC) is the most common women cancer and cause of cancer death. Despite decades of scientific progress in BC treatments, the clinical benefit of new drugs is modest in several cases. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway mutations are frequent in BC (20-40%) and are significant causes of aggressive tumor behavior, as well as treatment resistance. Improving knowledge of the PI3K/AKT/mTOR pathway is an urgent need. This review aims to highlight the central role of PI3K-mTORC1/C2 mutations in the different BC subtypes, in terms of clinical outcomes and treatment efficacy. The broad base of knowledge in tumor biology is a key point for personalized BC therapy in the precision medicine era.
Collapse
Affiliation(s)
- Krisida Cerma
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Federico Piacentini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
| | - Luca Moscetti
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
| | - Monica Barbolini
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
| | - Fabio Canino
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Antonio Tornincasa
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Federica Caggia
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Sara Cerri
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Alessia Molinaro
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, 41122 Modena, Italy
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
| | - Claudia Omarini
- Division of Medical Oncology, University Hospital of Modena, 41122 Modena, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
- Correspondence: ; Tel.: +39-059-422-2845
| |
Collapse
|
23
|
Hanan EJ, Braun MG, Heald RA, MacLeod C, Chan C, Clausen S, Edgar KA, Eigenbrot C, Elliott R, Endres N, Friedman LS, Gogol E, Gu XH, Thibodeau RH, Jackson PS, Kiefer JR, Knight JD, Nannini M, Narukulla R, Pace A, Pang J, Purkey HE, Salphati L, Sampath D, Schmidt S, Sideris S, Song K, Sujatha-Bhaskar S, Ultsch M, Wallweber H, Xin J, Yeap S, Young A, Zhong Y, Staben ST. Discovery of GDC-0077 (Inavolisib), a Highly Selective Inhibitor and Degrader of Mutant PI3Kα. J Med Chem 2022; 65:16589-16621. [PMID: 36455032 DOI: 10.1021/acs.jmedchem.2c01422] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Small molecule inhibitors that target the phosphatidylinositol 3-kinase (PI3K) signaling pathway have received significant interest for the treatment of cancers. The class I isoform PI3Kα is most commonly associated with solid tumors via gene amplification or activating mutations. However, inhibitors demonstrating both PI3K isoform and mutant specificity have remained elusive. Herein, we describe the optimization and characterization of a series of benzoxazepin-oxazolidinone ATP-competitive inhibitors of PI3Kα which also induce the selective degradation of the mutant p110α protein, the catalytic subunit of PI3Kα. Structure-based design informed isoform-specific interactions within the binding site, leading to potent inhibitors with greater than 300-fold selectivity over the other Class I PI3K isoforms. Further optimization of pharmacokinetic properties led to excellent in vivo exposure and efficacy and the identification of clinical candidate GDC-0077 (inavolisib, 32), which is now under evaluation in a Phase III clinical trial as a treatment for patients with PIK3CA-mutant breast cancer.
Collapse
Affiliation(s)
- Emily J Hanan
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Robert A Heald
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - Calum MacLeod
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - Connie Chan
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Saundra Clausen
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Kyle A Edgar
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Charles Eigenbrot
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Elliott
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - Nicholas Endres
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lori S Friedman
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Emily Gogol
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xiao-Hui Gu
- Pharmaron Beijing Co., Ltd, No. 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | | | - Philip S Jackson
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - James R Kiefer
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jamie D Knight
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - Michelle Nannini
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Raman Narukulla
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - Amanda Pace
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jodie Pang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hans E Purkey
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Laurent Salphati
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Deepak Sampath
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Stephen Schmidt
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steve Sideris
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Kyung Song
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Mark Ultsch
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Heidi Wallweber
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jianfeng Xin
- Pharmaron Beijing Co., Ltd, No. 6 Taihe Road, BDA, Beijing 100176, P. R. China
| | - SiewKuen Yeap
- Early Discovery Charles River, 7/8 Spire Green Centre, Flex Meadow, Harlow, Essex CM19 5TR, U.K
| | - Amy Young
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yu Zhong
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steven T Staben
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
24
|
Leung M, Rodrigues P, Roitman D. Ketoacidosis in a Patient with Type 2 Diabetes Requiring Alpelisib: Learnings and Observations Regarding Alpelisib Initiation and Rechallenge. Onco Targets Ther 2022; 15:1309-1315. [PMID: 36330532 PMCID: PMC9624212 DOI: 10.2147/ott.s370244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/09/2022] [Indexed: 11/06/2022] Open
Abstract
Background Diabetic ketoacidosis (DKA) is a rare complication of alpelisib, but cases of DKA are reported. Alpelisib's safety in patients with long-standing, suboptimally controlled diabetes is unclear since clinical trials of alpelisib did not include them. Case A case is presented on a patient with metastatic breast cancer and type 2 diabetes admitted for DKA eleven days after starting alpelisib. Since DKA is implicated in antihyperglycemics that inhibit sodium-glucose cotransporter-2 (SGLT2) inhibitors, her empagliflozin was discontinued. Alpelisib was also held since it was recently initiated. After the DKA resolved, she was discharged and restarted alpelisib. Within 4 hours of taking the first dose, the patient developed a second episode of DKA, and alpelisib treatment was stopped permanently. Conclusion Patients with long-standing type 2 diabetes are at high risk of alpelisib-induced Grade 3 and 4 hyperglycemia, including DKA. It is essential to communicate with non-oncology stakeholders about the risk of DKA with alpelisib as it can be overlooked for more common causes. Restarting alpelisib can result in severe hyperglycemia or DKA within 24 hours of the first dose. In this population, the risks associated with rechallenging alpelisib must be heavily weighed against its benefits. Before restarting alpelisib, a thorough evaluation of the appropriateness of the patient's antihyperglycemics and diet must occur to anticipate and mitigate a second event. Antihyperglycemics independent of the PI3K/AKT/mTOR pathway may be preferred agents. A plan should be in place to quickly respond to rising glycemia and early referral to a diabetologist or endocrinologist is recommended. Continuous glucose monitoring and hospital admission are recommended during rechallenge. A better understanding of alpelisib-induced hyperglycemia, especially in patients with diabetes, is required to navigate alpelisib treatment safely. Emphasis should be placed on patient education of symptoms and monitoring parameters.
Collapse
Affiliation(s)
- Mova Leung
- Cancer Care Program, North York General Hospital, Toronto, Ontario, Canada,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada,Correspondence: Mova Leung, Email
| | - Paulina Rodrigues
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Daryl Roitman
- Cancer Care Program, North York General Hospital, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Mellinghoff IK, Cloughesy TF. Balancing Risk and Efficiency in Drug Development for Rare and Challenging Tumors: A New Paradigm for Glioma. J Clin Oncol 2022; 40:3510-3519. [PMID: 35201903 PMCID: PMC10166355 DOI: 10.1200/jco.21.02166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 12/14/2022] Open
Abstract
The process of developing cancer therapies is well established and has enabled the incorporation of many new drugs and classes of agents into the standard of care for common cancers. Clinical drug development is fundamentally different for rare and difficult-to-treat solid tumors, such as glioma or pancreatic cancer. The failure to develop effective new agents for the latter diseases has discouraged the development of therapeutics for these cancers. Using glioma as an example, we describe a process toward obtaining more reliable early-stage signals of drug activity and a process toward translating those signals into clinical benefits with more efficient late-stage development. If linked together, these processes should increase the likelihood of benefit in late-stage settings at a lower cost and encourage more drug development for patients with rare and difficult-to-treat cancers.
Collapse
Affiliation(s)
- Ingo K. Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timothy F. Cloughesy
- Department of Neurology, Ronald Reagan UCLA Medical Center, University of California, Los Angeles, CA
| |
Collapse
|
26
|
Jfri A, Meltzer R, Mostaghimi A, LeBoeuf N, Guggina L. Incidence of Cutaneous Adverse Events With Phosphoinositide 3-Kinase Inhibitors as Adjuvant Therapy in Patients With Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2022; 8:2797488. [PMID: 36227613 PMCID: PMC9562095 DOI: 10.1001/jamaoncol.2022.4327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/27/2022] [Indexed: 11/14/2022]
Abstract
Importance The phosphoinositide 3-kinase (PI3K) pathway is among the most frequently activated pathways in human cancers. As the use of PI3K inhibitors for cancer treatment grows, there is increasing need for understanding the cutaneous effects associated with these therapies. Objective To systematically review the published literature reporting incidence of cutaneous adverse events with PI3K inhibitors and to provide pooled incidence estimates using meta-analysis. Data Sources This systematic review and meta-analysis was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines. The literature search concerned entries through September 2021 in the following sources: PubMed, Cochrane registry, ClinicalTrials.gov, and evidence from the NHS UK and Trip medical database. To analyze PI3K inhibitors' cutaneous adverse events incidence, only randomized clinical trials (RCTs) were considered. The search strategy used the following keywords: (prevalence OR incidence OR epidemiology) and (phosphoinositide 3 kinase inhibitors OR PI3K inhibitors). No language restriction was applied. Analysis was conducted on July 1, 2022. Study Selection Studies included phase 2 and phase 3 RCTs that reported incidence of cutaneous adverse events associated with use of PI3K inhibitors. Data Extraction and Measures Data extracted included sex, medication name and class, sample size, rash incidence, and grade. The bias risk was assessed by the Cochrane tool for risk of bias assessment in RCTs. Main Outcomes and Measures The primary outcome was incidence of PI3K inhibitor cutaneous adverse events (with 95% CIs) among the overall population and among subgroups. Between-study heterogeneity was assessed using the I2 statistic. Results The analysis found the incidence of PI3K inhibitor cutaneous events of any grade to be 29.30% in the intervention group, translating to a pooled odds ratio (OR) for incidence of cutaneous adverse events of any grades of 2.55 (95% CI, 1.74-3.75). Incidence of severe grade (grade ≥3) of rash in the intervention group was estimated to be 6.95%, yielding a pooled Peto OR of 4.64 (95% CI, 2.70-7.97). Subgroup analyses revealed that the incidence of severe cutaneous adverse events (grade ≥3) was higher with the use of Pan-class-1 PI3K inhibitors (OR, 6.67; 95% CI, 4.28-10.38) than isoform-selective PI3K inhibitors (OR, 6.37; 95% CI, 3.25-12.48). Conclusions and Relevance This systematic review and meta-analysis identified an overall incidence of PI3K inhibitor cutaneous adverse events of any grade to be 29.30% with a pooled OR of 2.55; (95% CI, 1.74-3.75). These findings clarify the risk of cutaneous adverse events associated with this important class of anticancer therapies.
Collapse
Affiliation(s)
- Abdulhadi Jfri
- Harvard Medical School, Boston, Massachusetts
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rachel Meltzer
- Harvard Medical School, Boston, Massachusetts
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Arash Mostaghimi
- Harvard Medical School, Boston, Massachusetts
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Nicole LeBoeuf
- Harvard Medical School, Boston, Massachusetts
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | |
Collapse
|
27
|
Xu H, Wang Y, Han Y, Wu Y, Wang J, Xu B. CDK4/6 inhibitors versus PI3K/AKT/mTOR inhibitors in women with hormone receptor-positive, HER2-negative metastatic breast cancer: An updated systematic review and network meta-analysis of 28 randomized controlled trials. Front Oncol 2022; 12:956464. [PMID: 36091147 PMCID: PMC9449843 DOI: 10.3389/fonc.2022.956464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Updated evidence was required to compare the efficacy and safety of cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) inhibitors for patients with hormone receptor-positive and HER2-negative metastatic breast cancer. Methods A systematic review and network meta-analysis was conducted utilizing data from randomized controlled trials (RCTs) that contained interventions of CDK4/6 inhibitors or PI3K/AKT/mTOR inhibitors. Progression-free survival (PFS), overall survival (OS), and treatment-related adverse events (TRAEs) were primary outcomes of interest. Pooled hazard ratios (HRs) and odds ratios (ORs) with 95% credible intervals (CrIs) were used to assess the survival outcomes and safety profiles, respectively. Results A total of 28 RCTs with 12,129 participants were included. Pooled analysis showed that CDK4/6 inhibitors significantly prolonged PFS than PI3K/AKT/mTOR inhibitors (HR, 0.81; 95% CrI, 0.69–0.94), whereas no significant differences were detected regarding OS. After balancing the treatment lines and metastatic sites, the superiority of CDK4/6 inhibitors only appeared in the visceral and non-visceral subgroups. Among CDK4/6 inhibitors, abemaciclib was significantly better than others in ≥3 grade neutropenia (OR, 0.04; 95% CrI, 0.01–0.15). The incidence of stomatitis and digestive disorders was different among diverse kinds of PI3K/AKT/mTOR inhibitors. Discrepancies appeared regarding TRAEs of hepatotoxicity, diarrhea, and hyperglycemia among different interventions. Conclusions CDK4/6 inhibitors showed better efficacy in PFS, but the benefits disappeared when taking treatment line into consideration. Specific and discrepant safety profiles were found in two categories of agents. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO, identifier CRD42022321172.
Collapse
Affiliation(s)
| | | | | | | | - Jiayu Wang
- *Correspondence: Binghe Xu, ; Jiayu Wang,
| | - Binghe Xu
- *Correspondence: Binghe Xu, ; Jiayu Wang,
| |
Collapse
|
28
|
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: 0.5] [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
|
29
|
Predictive biomarkers for molecularly targeted therapies and immunotherapies in breast cancer. Arch Pharm Res 2022; 45:597-617. [PMID: 35982262 DOI: 10.1007/s12272-022-01402-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Globally, breast cancer is the most common malignancy in women. Substantial efforts have been made to develop novel therapies, including targeted therapies and immunotherapies, for patients with breast cancer who do not respond to standard therapies. Consequently, new targeted therapies, such as cyclin-dependent kinase 4 and 6 inhibitors, poly (ADP-ribose) polymerase inhibitors, phosphoinositide 3-kinase inhibitor, and antibody-drug conjugates targeting human epidermal growth factor receptor 2 or trophoblast cell surface antigen-2, and immune checkpoint inhibitor targeting programmed cell death-1, have been developed and are now in clinical use. However, only some patients have benefited from these novel therapies; therefore, the identification and validation of reliable or more accurate biomarkers for predicting responses to these agents remain a major challenge. This review summarizes the currently available predictive biomarkers for breast cancer and describes recent efforts undertaken to identify potential predictive markers for molecularly targeted therapies and immune checkpoint inhibitors.
Collapse
|
30
|
Zhu K, Wu Y, He P, Fan Y, Zhong X, Zheng H, Luo T. PI3K/AKT/mTOR-Targeted Therapy for Breast Cancer. Cells 2022; 11:2508. [PMID: 36010585 PMCID: PMC9406657 DOI: 10.3390/cells11162508] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 12/25/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB/AKT) and mechanistic target of rapamycin (mTOR) (PAM) pathways play important roles in breast tumorigenesis and confer worse prognosis in breast cancer patients. The inhibitors targeting three key nodes of these pathways, PI3K, AKT and mTOR, are continuously developed. For breast cancer patients to truly benefit from PAM pathway inhibitors, it is necessary to clarify the frequency and mechanism of abnormal alterations in the PAM pathway in different breast cancer subtypes, and further explore reliable biomarkers to identify the appropriate population for precision therapy. Some PI3K and mTOR inhibitors have been approved by regulatory authorities for the treatment of specific breast cancer patient populations, and many new-generation PI3K/mTOR inhibitors and AKT isoform inhibitors have also been shown to have good prospects for cancer therapy. This review summarizes the changes in the PAM signaling pathway in different subtypes of breast cancer, and the latest research progress about the biomarkers and clinical application of PAM-targeted inhibitors.
Collapse
Affiliation(s)
- Kunrui Zhu
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanqi Wu
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ping He
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yu Fan
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Xiaorong Zhong
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Hong Zheng
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Ting Luo
- Breast Disease Center, Cancer Center, West China Hospital, Sichuan University, Chengdu 610000, China
- Multi-Omics Laboratory of Breast Diseases, State Key Laboratory of Biotherapy, National Collaborative, Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610000, China
| |
Collapse
|
31
|
Lai HZ, Han JR, Fu X, Ren YF, Li ZH, You FM. Targeted Approaches to HER2-Low Breast Cancer: Current Practice and Future Directions. Cancers (Basel) 2022; 14:cancers14153774. [PMID: 35954438 PMCID: PMC9367369 DOI: 10.3390/cancers14153774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary HER2-low breast cancer (BC) accounts for more than half of breast cancer patients. Anti-HER2 therapy has been ineffective in HER2-low BC, for which palliative chemotherapy is the main treatment modality. The definitive efficacy of T-Dxd in HER2-low BC breaks previous treatment strategies, which will redefine HER2-low and thus reshape anti-HER2 therapy. This review summarizes detection technologies and novel agents for HER2-low BC, and explores their possible role in future clinics, to provide ideas for the diagnosis and treatment of HER2-low BC. Abstract HER2-low breast cancer (BC) has a poor prognosis, making the development of more suitable treatment an unmet clinical need. While chemotherapy is the main method of treatment for HER2-low BC, not all patients benefit from it. Antineoplastic therapy without chemotherapy has shown promise in clinical trials and is being explored further. As quantitative detection techniques become more advanced, they assist in better defining the expression level of HER2 and in guiding the development of targeted therapies, which include directly targeting HER2 receptors on the cell surface, targeting HER2-related intracellular signaling pathways and targeting the immune microenvironment. A new anti-HER2 antibody-drug conjugate called T-DM1 has been successfully tested and found to be highly effective in clinical trials. With this progress, it could eventually be transformed from a disease without a defined therapeutic target into a disease with a defined therapeutic molecular target. Furthermore, efforts are being made to compare the sequencing and combination of chemotherapy, endocrine therapy, and HER2-targeted therapy to improve prognosis to customize the subtype of HER2 low expression precision treatment regimens. In this review, we summarize the current and upcoming treatment strategies, to achieve accurate management of HER2-low BC.
Collapse
|
32
|
Verma V, Bhari N. Ribociclib Induced Photosensitive Skin Lesions. Indian Dermatol Online J 2022; 14:105-106. [PMID: 36776167 PMCID: PMC9910512 DOI: 10.4103/idoj.idoj_99_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
| | - Neetu Bhari
- Department of Dermatology, AIIMS, New Delhi, India,Address for correspondence: Dr. Neetu Bhari, All India Institute of Medical Sciences, New Delhi, India. E-mail:
| |
Collapse
|
33
|
PI3K Inhibitor Eruptions: an Overview of Diagnostic and Management Strategies for the Inpatient Dermatologist. CURRENT DERMATOLOGY REPORTS 2022. [DOI: 10.1007/s13671-022-00365-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
34
|
Stamou MI, Chen C, Wander SA, Supko JG, Juric D, Bardia A, Wexler DJ. Severe Lactic Acidosis Complicated by Insulin-Resistant Hyperosmolar Hyperglycemic Syndrome in a Patient With Metastatic Breast Cancer Undergoing AKT-Inhibitor Therapy. JCO Precis Oncol 2022; 6:e2100428. [PMID: 35700410 PMCID: PMC9384915 DOI: 10.1200/po.21.00428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/23/2022] [Accepted: 05/02/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Maria I. Stamou
- Endocrine Division, Massachusetts General Hospital, Boston, MA
| | - Christopher Chen
- Department of Medicine, Stanford University School of Medicine,Palo Alto, CA
| | - Seth A. Wander
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA
| | - Jeffrey G. Supko
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA
| | - Dejan Juric
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA
| | - Aditya Bardia
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA
| | - Deborah J. Wexler
- Harvard Medical School, Boston, MA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
35
|
The History of Early Breast Cancer Treatment. Genes (Basel) 2022; 13:genes13060960. [PMID: 35741721 PMCID: PMC9222657 DOI: 10.3390/genes13060960] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
“The story of cancer is the story of human ingenuity, resilience, and perseverance, but also of hubris, paternalism, and misperception” (Siddhartha Mukherjee). The present review discusses the evolution of early breast cancer (BC) treatment philosophy in the last 50 years and the shift from an emphasis on local therapy to an emphasis on systemic precision treatment options.
Collapse
|
36
|
Gadkar K, Friedrich C, Hurez V, Ruiz M, Dickmann L, Kumar Jolly M, Schutt L, Jin J, Ware JA, Ramanujan S. Quantitative systems pharmacology model-based investigation of adverse gastrointestinal events associated with prolonged treatment with PI3-kinase inhibitors. CPT Pharmacometrics Syst Pharmacol 2022; 11:616-627. [PMID: 34850607 PMCID: PMC9124351 DOI: 10.1002/psp4.12749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 10/06/2021] [Accepted: 11/09/2021] [Indexed: 11/25/2022] Open
Abstract
Several PI3K inhibitors are in clinical development for the treatment of various forms of cancers, including pan-PI3K inhibitors targeting all four PI3K isoforms (α, β, γ, and δ), and isoform-selective inhibitors. Diarrhea and immune-mediated colitis are among the adverse events observed with PI3K inhibition which limits the maximal tolerated dose. A quantitative systems pharmacology model was developed to investigate PI3K-inhibitor-induced colitis. The effects of individual PI3K isoforms on relevant cellular pathways were incorporated into a mechanistic representation of mucosal inflammation. A virtual clinical population captures the observed clinical variability in the onset timing and rates of diarrhea and colitis for seven clinically tested PI3K inhibitors. Model-based analysis suggests that colitis development is governed by both the inhibition of PI3Kδ, which drives T cell differentiation and proliferation, and PI3Kα, which regulates epithelial barrier integrity. Specifically, when PI3Kα is inhibited below a given threshold, epithelial barrier dysfunction precipitates an exaggerated T effector response due to PI3Kδ-inhibition, leading to risk of diarrhea and colitis. This synergy explains why the lowest diarrhea and colitis rates are seen with the weakest PI3Kδ inhibition (alpelisib), and higher rates are seen with strong PI3Kδ inhibition if PI3Kα is even mildly inhibited (e.g., idelalisib), whereas strong PI3Kδ inhibition in the absence of PI3Kα inhibition does not result in high colitis rates (umbralisib). Thus, the model-based analysis suggests that PI3Kα and δ inhibition play unique but synergistic roles in driving colitis. Finally, we explore if and how dose-regimen might influence colitis rates for molecules that inhibit both PI3Kα and PI3Kδ.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jin Jin
- GenentechSouth San FranciscoCaliforniaUSA
| | | | | |
Collapse
|
37
|
Dong M, Shan B, Han X, Zhao X, Wang F, Zhu L, Ou Q, Ma X, Pan Y. Baseline Mutations and Up-Regulation of PI3K-AKT Pathway Serve as Potential Indicators of Lack of Response to Neoadjuvant Chemotherapy in Stage II/III Breast Cancer. Front Oncol 2022; 11:784985. [PMID: 35480699 PMCID: PMC9036956 DOI: 10.3389/fonc.2021.784985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Background Neoadjuvant chemotherapy (NAC) has been expanded to hormone receptor (HR) positive breast cancer (BC) patients with operable disease, to increase the likelihood of breast-conserving surgery. Genomic profiling at baseline would reveal NAC response relevant genomic features and signaling pathways, guiding clinical NAC utilization based on patients’ genomic characteristics. Methods We prospectively studied stage II/III BC patients who were eligible for breast-conserving surgery. Patients received epirubicin and cyclophosphamide for 4 cycles, followed by another 4-cycle docetaxel, and human epidermal growth factor receptor (HER2) positive patients were additionally treated with herceptin when using docetaxel (EC-T(H)). NAC responses were evaluated as pathologic complete response (pCR) or non-pathologic complete response (non-pCR). Genomic features related to NAC responses were identified by profiling baseline tumor tissues sampled one day before NAC, using whole-exome sequencing. Differentially expressed genes and up-/down-regulated pathways were investigated by performing RNA-sequencing. Results A total of 25 stage II/III BC patients were enrolled, including 5 patients ultimately evaluated as pCR and 20 patients evaluated as non-pCR. PIK3CA (48%) and TP53 (40%) mutations were enriched in patients not achieving pCR. Mutated phosphatidylinositol-3-kinase-AKT (PI3K-AKT) pathway and homologous recombinational repair pathway were also more frequently observed in patients evaluated as non-pCR. Significant arm-level amplifications (8q24.23 and 17q12) and deletions (1p32.2, 4p14, 7q11.23, 10q21.3, 11q23.3, etc.) were identified among patients not achieving pCR, while patients achieving pCR displayed no significant copy number alterations. Significantly up-regulated expression of PI3K-AKT pathway genes was also detected among patients failed to achieve pCR, compared to patients achieving pCR. Conclusion Compared to BC patients achieving pCR to NAC, aberrant activation of PI3K-AKT pathway genes were more frequently observed in patients not achieving pCR, consistent with the significant up-regulation of PI3K-AKT pathway gene expression in the non-pCR subgroup. Together, these findings indicate that upregulated PI3K-AKT pathway serves as a potential indicator of lack of response to NAC in stage II/III BC patients, and other effective therapeutic options are urgently needed for those resistant patients.
Collapse
Affiliation(s)
- Menghao Dong
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiaotian Zhao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Fufeng Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Liuqing Zhu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Xiaopeng Ma
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
38
|
Skolariki A, D’Costa J, Little M, Lord S. Role of PI3K/Akt/mTOR pathway in mediating endocrine resistance: concept to clinic. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:172-199. [PMID: 36046843 PMCID: PMC9400772 DOI: 10.37349/etat.2022.00078] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/11/2022] [Indexed: 01/06/2023] Open
Abstract
The majority of breast cancers express the estrogen receptor (ER) and for this group of patients, endocrine therapy is the cornerstone of systemic treatment. However, drug resistance is common and a focus for breast cancer preclinical and clinical research. Over the past 2 decades, the PI3K/Akt/mTOR axis has emerged as an important driver of treatment failure, and inhibitors of mTOR and PI3K are now licensed for the treatment of women with advanced ER-positive breast cancer who have relapsed on first-line hormonal therapy. This review presents the preclinical and clinical data that led to this new treatment paradigm and discusses future directions.
Collapse
Affiliation(s)
- Aglaia Skolariki
- Department of Oncology, University of Oxford, Churchill Hospital, OX3 7LE Oxford, UK
| | - Jamie D’Costa
- Department of Oncology, University of Oxford, Churchill Hospital, OX3 7LE Oxford, UK
| | - Martin Little
- Department of Oncology, Churchill Hospital, OX3 7LE Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Churchill Hospital, OX3 7LE Oxford, UK
| |
Collapse
|
39
|
Aebi S, Karlsson P, Wapnir IL. Locally advanced breast cancer. Breast 2021; 62 Suppl 1:S58-S62. [PMID: 34930650 PMCID: PMC9097810 DOI: 10.1016/j.breast.2021.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/05/2021] [Accepted: 12/12/2021] [Indexed: 11/28/2022] Open
Abstract
Locally advanced breast cancer (LABC) is defined here as inoperable breast adenocarcinoma without distant metastases. Patients with LABC require a multidisciplinary approach. Given the risk of distant metastasis, staging exams are necessary. The incidence of LABC (stages IIIB and IIIC) has decreased in recent years. LABC has rarely been investigated separately: patients with LABC have participated both in clinical trials of palliative and of neoadjuvant therapy. Most trials did not analyze responses and long-term outcomes independently; thus, the treatment of patients with LABC is extrapolated from studies of patients with less or more advanced disease. Pathologic confirmation and molecular profiling are essential for the choice of neoadjuvant chemotherapy. Preoperative endocrine therapy may be considered in certain clinical situations; the addition of a CDK4/6 inhibitor is being investigated. HER2 positive LABCs are targeted with anti-HER2 agents combined with chemotherapy. PD-1 and PD-L1 antibodies in ‘triple-negative’ LABC are promising. Excellent responses to neoadjuvant therapy enable conservative surgery in many patients; however, inflammatory breast cancer may still indicate mastectomy. Postoperative radiotherapy is usually indicated. Target volumes include breast/chest wall, axillary, supraclavicular and internal mammary nodal basins. Preoperative radiation therapy can be useful in patients without response to systemic therapies. Palliative surgery for poor responders after neoadjuvant systemic and radiation therapy can be considered. Multidisciplinary teams can optimize local control and prevent relapses. However, modest improvement in survival was achieved between 2000 and 2014 underscoring the unmet need in patients with LABC who will benefit from specific research efforts in this disease entity.
Collapse
Affiliation(s)
- Stefan Aebi
- Lucerne Cantonal Hospital and University of Bern, Cancer Center, Division of Medical Oncology, 6000, Lucerne 16, Switzerland.
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden.
| | - Irene L Wapnir
- Stanford University, Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA, 94305, USA.
| |
Collapse
|
40
|
Cusano E, Wong C, Taguedong E, Vaska M, Abedin T, Nixon N, Karim S, Tang P, Heng DYC, Ezeife D. Impact of Value Frameworks on the Magnitude of Clinical Benefit: Evaluating a Decade of Randomized Trials for Systemic Therapy in Solid Malignancies. Curr Oncol 2021; 28:4894-4928. [PMID: 34898590 PMCID: PMC8628676 DOI: 10.3390/curroncol28060412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 11/23/2022] Open
Abstract
In the era of rapid development of new, expensive cancer therapies, value frameworks have been developed to quantify clinical benefit (CB). We assessed the evolution of CB since the 2015 introduction of The American Society of Clinical Oncology and The European Society of Medical Oncology value frameworks. Randomized clinical trials (RCTs) assessing systemic therapies for solid malignancies from 2010 to 2020 were evaluated and CB (Δ) in 2010–2014 (pre-value frameworks (PRE)) were compared to 2015–2020 (POST) for overall survival (OS), progression-free survival (PFS), response rate (RR), and quality of life (QoL). In the 485 studies analyzed (12% PRE and 88% POST), the most common primary endpoint was PFS (49%), followed by OS (20%), RR (12%), and QoL (6%), with a significant increase in OS and decrease in RR as primary endpoints in the POST era (p = 0.011). Multivariable analyses revealed significant improvement in ΔOS POST (OR 2.86, 95% CI 0.46 to 5.26, p = 0.02) while controlling for other variables. After the development of value frameworks, median ΔOS improved minimally. The impact of value frameworks has yet to be fully realized in RCTs. Efforts to include endpoints shown to impact value, such as QoL, into clinical trials are warranted.
Collapse
Affiliation(s)
- Ellen Cusano
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Correspondence:
| | - Chelsea Wong
- Faculty of Science, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Eddy Taguedong
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Marcus Vaska
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| | - Tasnima Abedin
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| | - Nancy Nixon
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| | - Safiya Karim
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| | - Patricia Tang
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| | - Daniel Y. C. Heng
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| | - Doreen Ezeife
- Tom Baker Cancer Centre, Calgary, AB T2N 4N2, Canada; (M.V.); (T.A.); (N.N.); (S.K.); (P.T.); (D.Y.C.H.); (D.E.)
| |
Collapse
|
41
|
Chen X, Wei L, Chi L, Guo X, Chen C, Guo Z, Liang J, Zheng Y, He J, Ye X. Adverse events of alpelisib: A postmarketing study of the World Health Organization pharmacovigilance database. Br J Clin Pharmacol 2021; 88:2180-2189. [PMID: 34786743 DOI: 10.1111/bcp.15143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/13/2021] [Accepted: 11/01/2021] [Indexed: 01/07/2023] Open
Abstract
AIMS To explore and describe the adverse reaction signals in the safety reporting for alpelisib. METHODS We performed a disproportionality analysis of the World Health Organization's VigiBase pharmacovigilance database from 1 January 2019 to 30 June 2021. Disproportionality analysis by information components (ICs) were used to evaluate the potential association between adverse events (AEs) and alpelisib. RESULTS A total of 33 327 reports were extracted, 5695 of them were chosen with alpelisib as the suspected drug. After combining the same ID, 687 cases remained. The 45-64-years group had the most cases (n = 203, 29.55%). There were 129 Preferred Terms with significant signals. Hyperglycaemia (IC025 = 6.74), breast cancer metastatic (IC025 = 5.85) and metastases to liver (IC025 = 4.70) were the AEs with the strongest signal. AEs with the most cases were hyperglycaemia (n = 595), rash (n = 535) and diarrhoea (n = 475). CONCLUSION We established a comprehensive list of AEs potentially associated with alpelisib. AEs with the most significant signals were hyperglycaemia, breast cancer metastatic, metastases to liver. The AEs with the most cases were hyperglycaemia, rash, diarrhoea, blood glucose increase and nausea.
Collapse
Affiliation(s)
| | | | | | | | - Chenxin Chen
- Department of Health Statistics, Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Zhijian Guo
- Department of Health Statistics, Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Jizhou Liang
- Department of Health Statistics, Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Yi Zheng
- Department of Health Statistics, Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Jia He
- Department of Health Statistics, Faculty of Health Service, Naval Medical University, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Faculty of Health Service, Naval Medical University, Shanghai, China
| |
Collapse
|
42
|
Nabieva N, Fasching PA. Endocrine Treatment for Breast Cancer Patients Revisited-History, Standard of Care, and Possibilities of Improvement. Cancers (Basel) 2021; 13:5643. [PMID: 34830800 PMCID: PMC8616153 DOI: 10.3390/cancers13225643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 01/16/2023] Open
Abstract
PURPOSE OF REVIEW Due to the findings of current studies and the approval of novel substances for the therapy of hormone-receptor-positive breast cancer patients, the established standards of endocrine treatment are changing. The purpose of this review is to give an overview of the history of endocrine treatment, to clarify its role in the present standard of care, and to discuss the possibilities of improvement. RECENT FINDINGS Tamoxifen, aromatase inhibitors, and fulvestrant are the main drugs that have been used for decades in the therapy of hormone-receptor-positive breast cancer patients. However, since a relevant number of women suffer at some point from disease recurrence or progression, several novel substances are being investigated to overcome resistance mechanisms by interfering with certain signaling pathways, such as the PI3K/AKT/mTOR or the CDK4/6 pathways. mTOR and CDK4/6 inhibitors were the first drugs approved for this purpose and many more are in development. SUMMARY Endocrine treatment is one of the best tolerable cancer therapies available. Continuous investigation serves to improve patients' outcomes and modernize the current standard of care. Considering the resistance mechanisms and substances analyzed against these, endocrine treatment of hormone-receptor-positive breast cancer is on the brink of a new era.
Collapse
Affiliation(s)
- Naiba Nabieva
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
- Novartis Oncology, Novartis Pharma GmbH, 90429 Nuremberg, Germany
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| |
Collapse
|
43
|
Wilson BE, Cescon DW. Accelerating drug access from advanced to early breast cancer: the special case of oral selective estrogen receptor degraders. Curr Opin Oncol 2021; 33:538-546. [PMID: 34555836 DOI: 10.1097/cco.0000000000000786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW For hormone receptor positive breast cancer, the development of endocrine resistance commonly occurs, presenting as either disease progression in the metastatic setting or recurrence during or following adjuvant endocrine therapy. Various mechanisms of resistance have been described. In order to reduce or overcome endocrine resistance, there has been substantial interest in developing potent and orally bioavailable selective estrogen receptor degraders (SERDs) for metastatic disease and select patients with early-stage estrogen receptor positive breast cancer. RECENT FINDINGS At least 11 oral SERDs have entered clinical development. We review current studies in both the metastatic and neoadjuvant/adjuvant setting and present the available evidence of benefit and toxicity for these novel agents. Further characterization of changes to tissue-based biomarkers such as estrogen receptor, progesterone receptor and Ki67 expression and blood-based biomarkers such as ctDNA and estrogen receptor 1 mutation may help to refine therapeutic strategies, combinations, and patient selection to identify women who are most likely to benefit from these novel endocrine agents. SUMMARY Although SERDs have clear therapeutic potential based on nonclinical studies and have demonstrated early signs of activity in phase I and II studies in the metastatic setting, ongoing research is needed to clarify when and in whom these agents may have greatest clinical benefit.
Collapse
Affiliation(s)
- Brooke E Wilson
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.,University of New South Wales, Kensington, New South Wales, Australia
| | - David W Cescon
- Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
44
|
Sella T, Weiss A, Mittendorf EA, King TA, Pilewskie M, Giuliano AE, Metzger-Filho O. Neoadjuvant Endocrine Therapy in Clinical Practice: A Review. JAMA Oncol 2021; 7:1700-1708. [PMID: 34499101 DOI: 10.1001/jamaoncol.2021.2132] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Importance In clinical practice, neoadjuvant endocrine therapy (NET) is rarely used despite being an effective treatment modality able to downstage tumors and facilitate breast-conserving surgery. Observations Using data from studies conducted since 2000, we provide readers with a critical in-depth review on clinical aspects related to the application of NET in the treatment of hormone receptor (HR)-positive/ERBB2 (formerly HER2)-negative breast cancer. This includes an overview of patient-selection criteria, regimen choice, treatment duration, evaluation of response by imaging, interpretation of pathology after treatment, and surgical considerations. Areas of controversy include the use of gene-expression tests for patient selection, treatment of premenopausal women, surgical management of the axilla after NET, and adjuvant systemic therapy decision-making, including the use of chemotherapy. Conclusions and Relevance NET is an optimal treatment modality for a considerable proportion of postmenopausal women diagnosed with HR-positive tumors. The treatment landscape for HR-positive breast cancer is evolving, with novel agents and the growing use of gene expression profiling to define treatment selection. As such, it is likely that NET use will increase and the practical considerations outlined here will become more important.
Collapse
Affiliation(s)
- Tal Sella
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | - Anna Weiss
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tari A King
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts.,Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Melissa Pilewskie
- Breast Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Armando E Giuliano
- Division of Surgical Oncology, Department of Surgery, Cedars-Sinai Health System, Los Angeles, California
| | - Otto Metzger-Filho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| |
Collapse
|
45
|
Kaboli PJ, Imani S, Jomhori M, Ling KH. Chemoresistance in breast cancer: PI3K/Akt pathway inhibitors vs the current chemotherapy. Am J Cancer Res 2021; 11:5155-5183. [PMID: 34765318 PMCID: PMC8569340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/12/2021] [Indexed: 06/13/2023] Open
Abstract
Breast cancer is the most prevalent type of cancer among women. Several types of drugs, targeting the specific proteins expressed on the breast cancer cell surface (such as receptor tyrosine kinases and immune checkpoint regulators) and proteins involved in cell cycle and motility (including cyclin-dependent kinases, DNA stabilisers, and cytoskeleton modulators) are approved for different subtypes of breast cancer. However, breast cancer also has a poor response to conventional chemotherapy due to intrinsic and acquired resistance, and an Akt fingerprint is detectable in most drug-resistant cases. Overactivation of Akt and its upstream and downstream regulators in resistant breast cancer cells is considered a major potential target for novel anti-cancer therapies, suggesting that Akt signalling acts as a cellular mechanism against chemotherapy. The present review has shown that sustained activation of Akt results in resistance to different types of chemotherapy. Akt signalling plays a cellular defence role against chemotherapy and (1) enhances multi-drug resistance, (2) increases reactive oxygen species at breast tumor microenvironment, (3) enhances anaerobic metabolism, (4) inhibits the tricarboxylic cycle, (5) promotes PD-L1 upregulation, (6) inhibits apoptosis, (7) increases glucose uptake, and more importantly (8) recruits and interconnects the plasma membrane, nucleus, endoplasmic reticulum, and mitochondria to hijack breast cancer cells and rescue these cells from chemotherapy. Therefore, Akt signalling is considered a cellular defence mechanism employed against chemotherapeutic effects. In addition, interfering roles of PI3K/Akt signalling on the current cytotoxic and molecularly targeted therapy as well as immunotherapy of breast cancer are discussed with a clinical approach. Although, alpelisib, a PIK3CA inhibitor, is the only PI3K/Akt pathway inhibitor approved for breast cancer, we also highlight well-evaluated inhibitors of PI3K/Akt signalling based on different subtypes of breast cancer, which are under clinical trials whether as monotherapy or in combination with other types of chemotherapy.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical UniversityTaichung 404, Taiwan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra MalaysiaSerdang, Selangor 43400, Malaysia
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical UniversityLuzhou, Sichuan 646000, P. R. China
| | - Masume Jomhori
- Department of Biotechnology Research, Razi Vaccine and Serum Research InstituteMashhad, Iran
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra MalaysiaSerdang, Selangor 43400, Malaysia
- Department of Genetics, Harvard Medical SchoolBoston, MA 02115, USA
| |
Collapse
|
46
|
Chandrasekaran S, Funk CR, Kleber T, Paulos CM, Shanmugam M, Waller EK. Strategies to Overcome Failures in T-Cell Immunotherapies by Targeting PI3K-δ and -γ. Front Immunol 2021; 12:718621. [PMID: 34512641 PMCID: PMC8427697 DOI: 10.3389/fimmu.2021.718621] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
PI3K-δ and PI3K-γ are critical regulators of T-cell differentiation, senescence, and metabolism. PI3K-δ and PI3K-γ signaling can contribute to T-cell inhibition via intrinsic mechanisms and regulation of suppressor cell populations, including regulatory T-cells and myeloid derived suppressor cells in the tumor. We examine an exciting new role for using selective inhibitors of the PI3K δ- and γ-isoforms as modulators of T-cell phenotype and function in immunotherapy. Herein we review the current literature on the implications of PI3K-δ and -γ inhibition in T-cell biology, discuss existing challenges in adoptive T-cell therapies and checkpoint blockade inhibitors, and highlight ongoing efforts and future directions to incorporate PI3K-δ and PI3K-γ as synergistic T-cell modulators in immunotherapy.
Collapse
Affiliation(s)
- Sanjay Chandrasekaran
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Christopher Ronald Funk
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Troy Kleber
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Chrystal M. Paulos
- Department of Surgery/Microbiology & Immunology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, United States
| |
Collapse
|
47
|
Burguin A, Diorio C, Durocher F. Breast Cancer Treatments: Updates and New Challenges. J Pers Med 2021; 11:808. [PMID: 34442452 PMCID: PMC8399130 DOI: 10.3390/jpm11080808] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) is the most frequent cancer diagnosed in women worldwide. This heterogeneous disease can be classified into four molecular subtypes (luminal A, luminal B, HER2 and triple-negative breast cancer (TNBC)) according to the expression of the estrogen receptor (ER) and the progesterone receptor (PR), and the overexpression of the human epidermal growth factor receptor 2 (HER2). Current BC treatments target these receptors (endocrine and anti-HER2 therapies) as a personalized treatment. Along with chemotherapy and radiotherapy, these therapies can have severe adverse effects and patients can develop resistance to these agents. Moreover, TNBC do not have standardized treatments. Hence, a deeper understanding of the development of new treatments that are more specific and effective in treating each BC subgroup is key. New approaches have recently emerged such as immunotherapy, conjugated antibodies, and targeting other metabolic pathways. This review summarizes current BC treatments and explores the new treatment strategies from a personalized therapy perspective and the resulting challenges.
Collapse
Affiliation(s)
- Anna Burguin
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada;
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
| | - Caroline Diorio
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
- Department of Preventive and Social Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada
| | - Francine Durocher
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1T 1C2, Canada;
- Cancer Research Center, CHU de Québec-Université Laval, Quebec City, QC G1V 4G2, Canada;
| |
Collapse
|
48
|
Chen X, Zhabyeyev P, Azad AK, Vanhaesebroeck B, Grueter CE, Murray AG, Kassiri Z, Oudit GY. Pharmacological and cell-specific genetic PI3Kα inhibition worsens cardiac remodeling after myocardial infarction. J Mol Cell Cardiol 2021; 157:17-30. [PMID: 33887328 DOI: 10.1016/j.yjmcc.2021.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND PI3Kα (Phosphoinositide 3-kinase α) regulates multiple downstream signaling pathways controlling cell survival, growth, and proliferation and is an attractive therapeutic target in cancer and obesity. The clinically-approved PI3Kα inhibitor, BYL719, is in further clinical trials for cancer and overgrowth syndrome. However, the potential impact of PI3Kα inhibition on the heart and following myocardial infarction (MI) is unclear. We aim to determine whether PI3Kα inhibition affects cardiac physiology and post-MI remodeling and to elucidate the underlying molecular mechanisms. METHODS AND RESULTS Wildtype (WT) 12-wk old male mice receiving BYL719 (daily, p.o.) for 10 days showed reduction in left ventricular longitudinal strain with normal ejection fraction, weight loss, mild cardiac atrophy, body composition alteration, and prolonged QTC interval. RNASeq analysis showed gene expression changes in multiple pathways including extracellular matrix remodeling and signaling complexes. After MI, both p110α and phospho-Akt protein levels were increased in human and mouse hearts. Pharmacological PI3Kα inhibition aggravated cardiac dysfunction and resulted in adverse post-MI remodeling, with increased apoptosis, elevated inflammation, suppressed hypertrophy, decreased coronary blood vessel density, and inhibited Akt/GSK3β/eNOS signaling. Selective genetic ablation of PI3Kα in endothelial cells was associated with worsened post-MI cardiac function and reduced coronary blood vessel density. In vitro, BYL719 suppressed Akt/eNOS activation, cell viability, proliferation, and angiogenic sprouting in coronary and human umbilical vein endothelial cells. Cardiomyocyte-specific genetic PI3Kα ablation resulted in mild cardiac systolic dysfunction at baseline. After MI, cardiac function markedly deteriorated with increased mortality concordant with greater apoptosis and reduced hypertrophy. In isolated adult mouse cardiomyocytes, BYL719 decreased hypoxia-associated activation of Akt/GSK3β signaling and cell survival. CONCLUSIONS PI3Kα is required for cell survival (endothelial cells and cardiomyocytes) hypertrophic response, and angiogenesis to maintain cardiac function after MI. Therefore, PI3Kα inhibition that is used as anti-cancer treatment, can be cardiotoxic, especially after MI.
Collapse
Affiliation(s)
- Xueyi Chen
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Pavel Zhabyeyev
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Abul K Azad
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | | - Chad E Grueter
- Division of Cardiovascular Medicine, Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
| | - Allan G Murray
- Department of Medicine, University of Alberta, Edmonton, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
49
|
Abstract
Approximately 70% of invasive breast cancers have some degree of dependence on the estrogen hormone for cell proliferation and growth. These tumors have estrogen and/or progesterone receptors (ER/PR+), generally referred to as hormone receptor positive (HR+) tumors, as indicated by the presence of positive staining and varying intensity levels of estrogen and/or progesterone receptors on immunohistochemistry. Therapies that inhibit ER signaling pathways, such as aromatase inhibitors (letrozole, anastrozole, exemestane), selective ER modulators (tamoxifen), and ER down-regulators (fulvestrant), are the mainstays of treatment for hormone-receptor-positive breast cancers. However, de novo or acquired resistance to ER targeted therapies is present in many tumors, leading to disease progression. The PI3K/AKT/mTOR pathway is implicated in sustaining endocrine resistance and has become the target of many new drugs for ER+ breast cancer. This article reviews the function of the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway and the various classes of PI3K pathway inhibitors that have been developed to disrupt this pathway signaling for the treatment of hormone-receptor-positive breast cancer.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/diagnosis
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors
- Class I Phosphatidylinositol 3-Kinases/genetics
- Class I Phosphatidylinositol 3-Kinases/metabolism
- DNA Mutational Analysis
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Humans
- Mutation
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Staging
- Phosphoinositide-3 Kinase Inhibitors/metabolism
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/antagonists & inhibitors
- Receptors, Progesterone/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Sara E Nunnery
- Breast Cancer Program, Division of Hematology/Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN, 37232-6307, USA
| | - Ingrid A Mayer
- Breast Cancer Program, Division of Hematology/Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center (VICC), Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN, 37232-6307, USA.
| |
Collapse
|
50
|
Tayyar Y, Idris A, Vidimce J, Ferreira DA, McMillan NAJ. Alpelisib and radiotherapy treatment enhances Alisertib-mediated cervical cancer tumor killing. Am J Cancer Res 2021; 11:3240-3251. [PMID: 34249458 PMCID: PMC8263691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/14/2021] [Indexed: 06/13/2023] Open
Abstract
Human papilloma virus (HPV) is the main causative agent in cervical cancers. High-risk HPV cancers, including cervical cancer, are driven by major HPV oncogene, E6 and E7, which promote uncontrolled cell growth and genomic instability. We have previously shown that the presence of HPV E7 sensitizes cells to inhibition of aurora kinases (AURKs), which regulates the control of cell entry into and through mitosis. Such treatment is highly effective at eliminating early tumors and reducing large, late tumors. In addition, the presence of HPV oncogenes also sensitizes cells to inhibition of phosphoinositide 3-kinases (PI3Ks), a family of enzymes involved in cellular functions such as cell growth and proliferation. Using MLN8237 (Alisertib), an oral, selective inhibitor of AURKs, we investigated whether Alisertib treatment can improve tumor response when combined with either radiotherapy (RT) treatment or with a PI3K inhibitor, BYL719 (Alpelisib). Indeed, both RT and Alpelisib significantly improved Alisertib-mediated tumor killing, and the promising achieved results warrant further development of these combinations, and potentially translating them to the clinics.
Collapse
|