151
|
Anaclerio F, Pilenzi L, Dell’Elice A, Ferrante R, Grossi S, Ferlito LM, Marinelli C, Gildetti S, Calabrese G, Stuppia L, Antonucci I. Clinical usefulness of NGS multi-gene panel testing in hereditary cancer analysis. Front Genet 2023; 14:1060504. [PMID: 37065479 PMCID: PMC10104445 DOI: 10.3389/fgene.2023.1060504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/04/2023] [Indexed: 02/04/2023] Open
Abstract
Introduction: A considerable number of families with pedigrees suggestive of a Mendelian form of Breast Cancer (BC), Ovarian Cancer (OC), or Pancreatic Cancer (PC) do not show detectable BRCA1/2 mutations after genetic testing. The use of multi-gene hereditary cancer panels increases the possibility to identify individuals with cancer predisposing gene variants. Our study was aimed to evaluate the increase in the detection rate of pathogenic mutations in BC, OC, and PC patients when using a multi-gene panel.Methods: 546 patients affected by BC (423), PC (64), or OC (59) entered the study from January 2020 to December 2021. For BC patients, inclusion criteria were i) positive cancer family background, ii) early onset, and iii) triple negative BC. PC patients were enrolled when affected by metastatic cancer, while OC patients were all submitted to genetic testing without selection. The patients were tested using a Next-Generation Sequencing (NGS) panel containing 25 genes in addition to BRCA1/2.Results: Forty-four out of 546 patients (8%) carried germline pathogenic/likely pathogenic variants (PV/LPV) on BRCA1/2 genes, and 46 (8%) presented PV or LPV in other susceptibility genes.Discussion: Our findings demonstrate the utility of expanded panel testing in patients with suspected hereditary cancer syndromes, since this approach increased the mutation detection rate of 15% in PC, 8% in BC and 5% in OC cases. In absence of multi-gene panel analysis, a considerable percentage of mutations would have been lost.
Collapse
Affiliation(s)
- Federico Anaclerio
- Center for Advanced Studies and Technology (CAST), G.d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Lucrezia Pilenzi
- Center for Advanced Studies and Technology (CAST), G.d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Anastasia Dell’Elice
- Center for Advanced Studies and Technology (CAST), G.d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Rossella Ferrante
- Center for Advanced Studies and Technology (CAST), G.d’Annunzio University of Chieti-Pescara, Chieti, Italy
- *Correspondence: Rossella Ferrante,
| | - Simona Grossi
- Eusoma Breast Centre, “G. Bernabeo” Hospital, Ortona, Italy
| | | | | | | | - Giuseppe Calabrese
- UOSD Genetica Oncoematologica, Dipartimento di Oncologico-Ematologico, Pescara, Italy
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), G.d’Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Ivana Antonucci
- Center for Advanced Studies and Technology (CAST), G.d’Annunzio University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
152
|
Wu HL, Luo ZY, He ZL, Gong Y, Mo M, Ming WK, Liu GY. All HER2-negative breast cancer patients need gBRCA testing: cost-effectiveness and clinical benefits. Br J Cancer 2023; 128:638-646. [PMID: 36564566 PMCID: PMC9938252 DOI: 10.1038/s41416-022-02111-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The OlympiA trial demonstrated the benefits of adjuvant usage of olaparib for high-risk patients with human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC) and germline BRCA (gBRCA) mutation. This provoked thoughts on the clinical criteria of gBRCA testing. This study aims to estimate the costs and benefits of gBRCA testing and adjuvant olaparib therapy for patients with triple-negative breast cancer (TNBC) and hormone-receptor (HR)-positive and HER2-negative BC in China and the United States of America (USA). METHODS We used a Markov chain decision tree analytic model to compare three gBRCA screening policies in China and the USA: (1) no gBRCA testing; (2) selected gBRCA testing and (3) universal gBRCA testing for nonmetastatic TNBC and HR-positive HER2-negative BC patients. We modelled the benefit of systemic therapy and risk-reducing surgeries among patients identified with pathogenic or likely pathogenic variants (PVs) in BRCA1 and BRCA2. RESULTS Changing from the selected gBRCA testing to the universal gBRCA testing in TNBC patients is cost-effective, with the incremental cost-effectiveness ratios (ICERs) being 10991.1 and 56518.2 USD/QALY in China and the USA, respectively. Expanding universal gBRCA testing to HR-positive HER2-negative BC and TNBC patients has ICERs of 2023.3 and 16611.1 USD/QALY in China and the USA, respectively. DISCUSSION By performing gBRCA testing on all HER2-negative BC patients, adjuvant olaparib can be offered to high-risk patients with a PV in BRCA1 or BRCA2. These patients are also candidates for risk-reducing surgeries, an important aspect of their survivorship care, and these interventions can improve survival outcomes. With the willingness-to-pay thresholds being 31,500.0 and 100,000.0 USD per QALY gained in China and the USA, respectively, universal gBRCA testing is likely cost-effective for all HER2-negative BC patients. This simplified criterion of gBRCA testing for BC is recommended for adoption by current guidelines in China and the USA.
Collapse
Affiliation(s)
- Huai-Liang Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zi-Yin Luo
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat‑sen University, Guangzhou, China
| | - Zong-Lin He
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Yue Gong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Miao Mo
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wai-Kit Ming
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, China
| | - Guang-Yu Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
153
|
Rodepeter FR, Teply-Szymanski J, Romey M, Grass A, Erber R, Lebeau A, Mack EKM, Tarawneh TS, Gremke N, Boekhoff J, Wündisch T, Wagner U, Jank P, Denkert C. [Clinically relevant molecular pathological diagnostics in breast cancer]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:39-49. [PMID: 36629894 DOI: 10.1007/s00292-022-01175-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/12/2023]
Abstract
In breast cancer, the current guideline for pathological workup includes recommendations for advanced molecular analysis of certain predictive molecular markers in addition to basic immunohistochemical diagnostics. These markers are determined depending on tumor stage, including sequencing techniques and immunohistochemical methods. This comprises the systematic investigation of molecular alterations such as PIK3CA or BRCA1,2 mutations, NTRK fusions, or microsatellite instability as a basis for targeted therapy. Further alterations, for example in the PI3K pathway, ESR1 alterations, or ERBB2 mutations, may also be relevant for individual therapy decisions especially in the context of resistant or relapsed disease. Thus, particularly in advanced stages, a more comprehensive molecular characterization of the tumor may reveal genetic alterations that act as tumor drivers and provide targets for personalized therapies. Due to the large number of potential molecular targets, NGS panel diagnostics are a suitable approach in this conjunction with immunohistochemical characterization and the individual clinical situation. Molecular based therapeutical strategies outside of entity-specific approvals should be discussed in an interdisciplinary team within the framework of a molecular tumor board.
Collapse
Affiliation(s)
- Fiona R Rodepeter
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Baldingerstr. 1, 35043, Marburg, Deutschland
| | - Julia Teply-Szymanski
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Baldingerstr. 1, 35043, Marburg, Deutschland
| | - Marcel Romey
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Baldingerstr. 1, 35043, Marburg, Deutschland
| | - Albert Grass
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Baldingerstr. 1, 35043, Marburg, Deutschland
| | - Ramona Erber
- Pathologisches Institut, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Deutschland
| | - Annette Lebeau
- Institut für Pathologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland.,Gemeinschaftspraxis für Pathologie, Lübeck, Deutschland
| | - Elisabeth K M Mack
- Abteilung für Hämatologie, Onkologie und Immunologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Marburg, Deutschland
| | - Thomas S Tarawneh
- Abteilung für Hämatologie, Onkologie und Immunologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Marburg, Deutschland
| | - Niklas Gremke
- Abteilung für Frauenheilkunde und Geburtshilfe, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Marburg, Deutschland
| | - Jelena Boekhoff
- Abteilung für Frauenheilkunde und Geburtshilfe, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Marburg, Deutschland
| | - Thomas Wündisch
- Comprehensive Cancer Center, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Marburg, Deutschland
| | - Uwe Wagner
- Abteilung für Frauenheilkunde und Geburtshilfe, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Marburg, Deutschland
| | - Paul Jank
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Baldingerstr. 1, 35043, Marburg, Deutschland
| | - Carsten Denkert
- Institut für Pathologie, Philipps-Universität Marburg und Universitätsklinikum Gießen und Marburg (UKGM), Baldingerstr. 1, 35043, Marburg, Deutschland.
| |
Collapse
|
154
|
Systemic Therapy for Hereditary Breast Cancers. Hematol Oncol Clin North Am 2023; 37:203-224. [PMID: 36435611 DOI: 10.1016/j.hoc.2022.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Approximately 5% to 10% of all breast cancers are hereditary; many of which are caused by pathogenic variants in genes required for homologous recombination, including BRCA1 and BRCA2. Here we discuss systemic treatment for such breast cancers, including approved chemotherapeutic approaches and also targeted treatment approaches using poly-(ADP ribose) polymerase inhibitors. We also discuss experimental approaches to treating hereditary breast cancer, including new small molecule DNA repair inhibitors and also immunomodulatory agents. Finally, we discuss how drug resistance emerges in patients with hereditary breast cancer, how this might be delayed or prevented, and how biomarker-adapted treatment is molding the future management of hereditary breast cancer.
Collapse
|
155
|
Zheng J, Li Z, Min W. Current status and future promise of next-generation poly (ADP-Ribose) polymerase 1-selective inhibitor AZD5305. Front Pharmacol 2023; 13:979873. [PMID: 36756144 PMCID: PMC9899804 DOI: 10.3389/fphar.2022.979873] [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: 06/28/2022] [Accepted: 12/19/2022] [Indexed: 01/24/2023] Open
Abstract
The family of poly (ADP-ribose) polymerases (PARPs) consists of 17 members, which have been demonstrated as having effects on a series of cellular processes, including DNA replication and repair. PARP inhibitors (PARPi) suppress DNA repair through "PARP trapping", thus, constitute an important treatment option for cancer nowadays. In addition, PARP inhibition and homologous recombination repair (HRR) defects are synthetically lethal, giving a promising therapeutic for homologous recombination repair deficient (HRD) tumors including BRCA mutation. However, overlapping hematologic toxicity causes PARPi to fail in combination with some first-line chemotherapies. Furthermore, recent literature has demonstrated that PARP1 inhibition and PARP1-DNA trapping are key for antitumor activity in HRD cancer models. Currently approved PARPi have shown varying levels of selectivity for the entire 17-member PARP family, hence contribute to toxicity. Together, these findings above have provided the necessity and feasibility of developing next-generation PARPi with improved selectivity for PARP1, expanding significant clinical values and wide application prospects both in monotherapy and combination with other anticancer agents. In this review, we summery the latest research of current approved PARPi, discuss the current status and future promise of next-generation PARP1-selective inhibitor AZD5305, including its reported progress up to now and anticipated impact on clinical.
Collapse
Affiliation(s)
- Jingcao Zheng
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Zhengyu Li
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China,*Correspondence: Zhengyu Li, ; Wenjiao Min,
| | - Wenjiao Min
- Psychosomatic Department, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China,*Correspondence: Zhengyu Li, ; Wenjiao Min,
| |
Collapse
|
156
|
Tsujino T, Takai T, Hinohara K, Gui F, Tsutsumi T, Bai X, Miao C, Feng C, Gui B, Sztupinszki Z, Simoneau A, Xie N, Fazli L, Dong X, Azuma H, Choudhury AD, Mouw KW, Szallasi Z, Zou L, Kibel AS, Jia L. CRISPR screens reveal genetic determinants of PARP inhibitor sensitivity and resistance in prostate cancer. Nat Commun 2023; 14:252. [PMID: 36650183 PMCID: PMC9845315 DOI: 10.1038/s41467-023-35880-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
Prostate cancer harboring BRCA1/2 mutations are often exceptionally sensitive to PARP inhibitors. However, genomic alterations in other DNA damage response genes have not been consistently predictive of clinical response to PARP inhibition. Here, we perform genome-wide CRISPR-Cas9 knockout screens in BRCA1/2-proficient prostate cancer cells and identify previously unknown genes whose loss has a profound impact on PARP inhibitor response. Specifically, MMS22L deletion, frequently observed (up to 14%) in prostate cancer, renders cells hypersensitive to PARP inhibitors by disrupting RAD51 loading required for homologous recombination repair, although this response is TP53-dependent. Unexpectedly, loss of CHEK2 confers resistance rather than sensitivity to PARP inhibition through increased expression of BRCA2, a target of CHEK2-TP53-E2F7-mediated transcriptional repression. Combined PARP and ATR inhibition overcomes PARP inhibitor resistance caused by CHEK2 loss. Our findings may inform the use of PARP inhibitors beyond BRCA1/2-deficient tumors and support reevaluation of current biomarkers for PARP inhibition in prostate cancer.
Collapse
Affiliation(s)
- Takuya Tsujino
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Tomoaki Takai
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kunihiko Hinohara
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fu Gui
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Takeshi Tsutsumi
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Xiao Bai
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Chenkui Miao
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Chao Feng
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Bin Gui
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Zsofia Sztupinszki
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Antoine Simoneau
- Department of Pathology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA
| | - Ning Xie
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Atish D Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
| | - Kent W Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute & Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Zoltan Szallasi
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Lee Zou
- Department of Pathology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA
| | - Adam S Kibel
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Li Jia
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
157
|
Abdel-Razeq H, Abujamous L, Al-Azzam K, Abu-Fares H, Bani Hani H, Alkyam M, Sharaf B, Elemian S, Tamimi F, Abuhijla F, Edaily S, Salama O, Abdulelah H, Daoud R, Abubaker M, Al-Atary A. Guideline-Based, Multi-Gene Panel Germline Genetic Testing for at-Risk Patients with Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:1-10. [PMID: 36660366 PMCID: PMC9844102 DOI: 10.2147/bctt.s394092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/15/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Genetic testing for at-risk patients with breast cancer should be routinely offered. Knowledge generated may influence both treatment decisions and cancer prevention strategies among the patients themselves and their relatives. In this study, we report on the prevalence and patterns of germline mutations, using commercially available next-generation sequencing (NGS)-based multi-gene panels (MGP). PATIENTS AND METHODS Consecutive at-risk breast cancer patients, as determined by international guidelines, were offered germline genetic testing using a 20-gene NGS-based panel at a reference lab. Samples of peripheral blood were obtained for DNA extraction and genetic variants were classified as benign/likely benign (negative), pathogenic/likely pathogenic (positive) or variants of uncertain significance (VUS). RESULTS A total of 1310 patients, median age (range) 43 (19-82) years, were enrolled. Age ≤45 years (n = 800, 61.1%) was the most common indication for testing. Positive family history of breast, ovarian, pancreatic or prostate cancers, and triple-negative disease were among the common indications. Among the whole group, 184 (14.0%) patients had pathogenic/likely pathogenic variants; only 90 (48.9%) were in BRCA1 or BRCA2, while 94 (51.9%) others had pathogenic variants in other genes; mostly in APC, TP53, CHEK2 and PALB2. Mutation rates were significantly higher among patients with positive family history (p = 0.009); especially if they were 50 years or younger at the time of breast cancer diagnosis (p < 0.001). Patients with triple-negative disease had relatively higher rate (17.5%), and mostly in BRCA1/2 genes (71.4%). Variants of uncertain significance (VUS) were reported in 559 (42.7%) patients; majority (90.7%) were in genes other than BRCA1 or BRCA2. CONCLUSION Pathogenic mutations in genes other than BRCA1/2 are relatively common and could have been missed if genetic testing was restricted to BRCA1/2. The significantly high rate of VUS associated with multi-gene panel testing can be disturbing.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Lama Abujamous
- Department of Cell Therapy & Applied Genomic, King Hussein Cancer Center, Amman, Jordan
| | - Khansa Al-Azzam
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hala Abu-Fares
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Mais Alkyam
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Baha’ Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Shatha Elemian
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Faris Tamimi
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Fawzi Abuhijla
- Department of Radiation Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Sarah Edaily
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Osama Salama
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hazem Abdulelah
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Rand Daoud
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Mohammad Abubaker
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Areej Al-Atary
- Department of Nursing, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
158
|
Schram AM, Colombo N, Arrowsmith E, Narayan V, Yonemori K, Scambia G, Zelnak A, Bauer TM, Jin N, Ulahannan SV, Colleoni M, Aftimos P, Donoghue MTA, Rosen E, Rudneva VA, Telli ML, Domchek SM, Galsky MD, Hoyle M, Chappey C, Stewart R, Blake-Haskins JA, Yap TA. Avelumab Plus Talazoparib in Patients With BRCA1/2- or ATM-Altered Advanced Solid Tumors: Results From JAVELIN BRCA/ATM, an Open-Label, Multicenter, Phase 2b, Tumor-Agnostic Trial. JAMA Oncol 2023; 9:29-39. [PMID: 36394867 PMCID: PMC9673021 DOI: 10.1001/jamaoncol.2022.5218] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/01/2022] [Indexed: 11/18/2022]
Abstract
Importance Nonclinical studies suggest that the combination of poly(ADP-ribose) polymerase and programmed cell death 1/programmed cell death-ligand 1 inhibitors has enhanced antitumor activity; however, the patient populations that may benefit from this combination have not been identified. Objective To evaluate whether the combination of avelumab and talazoparib is effective in patients with pathogenic BRCA1/2 or ATM alterations, regardless of tumor type. Design, Setting, and Participants In this pan-cancer tumor-agnostic phase 2b nonrandomized controlled trial, patients with advanced BRCA1/2-altered or ATM-altered solid tumors were enrolled into 2 respective parallel cohorts. The study was conducted from July 2, 2018, to April 12, 2020, at 42 institutions in 9 countries. Interventions Patients received 800 mg of avelumab every 2 weeks and 1 mg of talazoparib once daily. Main Outcomes and Measures The primary end point was confirmed objective response (OR) per RECIST 1.1 by blinded independent central review. Results A total of 200 patients (median [range] age, 59.0 [26.0-89.0] years; 132 [66.0%] women; 15 [7.5%] Asian, 11 [5.5%] African American, and 154 [77.0%] White participants) were enrolled: 159 (79.5%) in the BRCA1/2 cohort and 41 (20.5%) in the ATM cohort. The confirmed OR rate was 26.4% (42 patients, including 9 complete responses [5.7%]) in the BRCA1/2 cohort and 4.9% (2 patients) in the ATM cohort. In the BRCA1/2 cohort, responses were more frequent (OR rate, 30.3%; 95% CI, 22.2%-39.3%, including 8 complete responses [6.7%]) and more durable (median duration of response: 10.9 months [95% CI, 6.2 months to not estimable]) in tumor types associated with increased heritable cancer risk (ie, BRCA1/2-associated cancer types, such as ovarian, breast, prostate, and pancreatic cancers) and in uterine leiomyosarcoma (objective response in 3 of 3 patients and with ongoing responses greater than 24 months) compared with non-BRCA-associated cancer types. Responses in the BRCA1/2 cohort were numerically higher for patients with tumor mutational burden of 10 or more mutations per megabase (mut/Mb) vs less than 10 mut/Mb. The combination was well tolerated, with no new safety signals identified. Conclusions and Relevance In this phase 2b nonrandomized controlled trial, neither the BRCA1/2 nor ATM cohort met the prespecified OR rate of 40%. Antitumor activity for the combination of avelumab and talazoparib in patients with BRCA1/2 alterations was observed in some patients with BRCA1/2-associated tumor types and uterine leiomyosarcoma; benefit was minimal in non-BRCA-associated cancer types. Trial Registration ClinicalTrials.gov Identifier: NCT03565991.
Collapse
Affiliation(s)
| | - Nicoletta Colombo
- University of Milan-Bicocca and Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| | | | - Vivek Narayan
- Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia
| | - Kan Yonemori
- National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Todd M. Bauer
- Tennessee Oncology/Sarah Cannon Research Institute, Nashville
| | - Ning Jin
- Division of Medical Oncology, Wexner Medical Center, The Ohio State University, Columbus
| | | | - Marco Colleoni
- Division of Medical Senology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Philippe Aftimos
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Mark T. A. Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ezra Rosen
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vasilisa A. Rudneva
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Susan M. Domchek
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Matthew D. Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Ross Stewart
- now with Translational Medicine, Oncology at AstraZeneca, Cambridge, England, United Kingdom
- Pfizer, San Diego, California
| | | | - Timothy A. Yap
- The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
159
|
Albarrán V, Chamorro J, Pozas J, San Román M, Rosero DI, Saavedra C, Gion M, Cortés A, Escalera E, Guerra E, López Miranda E, Fernández Abad M, Martínez Jañez N. Maintained complete response to talazoparib in a BRCA-2 mutated metastatic luminal breast cancer: case report and review of literature. Front Oncol 2023; 13:1158981. [PMID: 37213307 PMCID: PMC10196382 DOI: 10.3389/fonc.2023.1158981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/20/2023] [Indexed: 05/23/2023] Open
Abstract
PARP inhibitors are progressively becoming a part of our therapeutic arsenal against BRCA-defective tumors, because of their capacity to induce synthetic lethality in cells with a deficiency in the homologous recombination repair system. Olaparib and talazoparib have been approved for metastatic breast cancer in carriers of germline BRCA mutations, which are found in approximately 6% of patients with breast cancer. We report the case of a patient with metastatic breast cancer, carrier of a germline mutation in BRCA2, with a complete response to first-line treatment with talazoparib, maintained after 6 years. To the best of our knowledge, this is the longest response reported with a PARP inhibitor in a BRCA-mutated tumor. We have made a review of literature, regarding the rationale for PARP inhibitors in carriers of BRCA mutations and their clinical relevance in the management of advanced breast cancer, as well as their emerging role in early stage disease, alone and in combination with other systemic therapies.
Collapse
|
160
|
Lynce F, Robson M. Clinical Use of PARP Inhibitors in BRCA Mutant and Non-BRCA Mutant Breast Cancer. Cancer Treat Res 2023; 186:91-102. [PMID: 37978132 DOI: 10.1007/978-3-031-30065-3_6] [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] [Indexed: 11/19/2023]
Abstract
The use of poly(ADP-ribose) polymerase (PARP) inhibitors for the treatment of patients with germline BRCA mutations (gBRCAm) and breast cancer, both in the early and advanced settings, is a success of genomically-directed treatment. These agents have been shown to be associated with longer progression-free survival when compared to standard chemotherapy, with an acceptable toxicity profile. A recent randomized trial demonstrated improved survival with the use of olaparib for 2 years compared to placebo in patients with early-stage high risk gBRCAm associated breast cancer. Ongoing research efforts are focused on identifying patients beyond those with BRCA1/2 or PALB2 mutations who may benefit from PARP inhibitors, exploring the overlapping mechanisms of resistance between platinum and PARP inhibitors and developing agents with less toxicity that will allow combinational strategies.
Collapse
Affiliation(s)
- Filipa Lynce
- Harvard Medical School, Medical Oncology, Dana-Farber Cancer Institute, Dana-Farber Brigham Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.
| | - Mark Robson
- Breast Medicine and Clinical Genetics Services, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 300 East 66th Street, Room 813, New York, NY, 10065, USA
| |
Collapse
|
161
|
Molecular Testing for Diagnostics, Prognostication, and Treatment Stratification in Cancers. Cancer J 2023; 29:3-8. [PMID: 36693151 DOI: 10.1097/ppo.0000000000000643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
ABSTRACT Precision cancer care, for essentially all cancer types, now requires molecular diagnostics to assess mutations, chromosomal alterations, and gene expression to personalize treatments for individual patients. Advances in the diagnostics and treatment options have moved the field forward from fundamental discoveries beginning in the 1960s to the development of many targeted therapies that can be as specific as targeting a single-base-pair mutation. Herein is a brief historical perspective on cancer precision medicine with current diagnostic, prognostic, and treatment stratification guidance for early- and late-stage cancers.
Collapse
|
162
|
Sokolova A, Johnstone KJ, McCart Reed AE, Simpson PT, Lakhani SR. Hereditary breast cancer: syndromes, tumour pathology and molecular testing. Histopathology 2023; 82:70-82. [PMID: 36468211 PMCID: PMC10953374 DOI: 10.1111/his.14808] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/12/2022] [Accepted: 09/18/2022] [Indexed: 12/09/2022]
Abstract
Hereditary factors account for a significant proportion of breast cancer risk. Approximately 20% of hereditary breast cancers are attributable to pathogenic variants in the highly penetrant BRCA1 and BRCA2 genes. A proportion of the genetic risk is also explained by pathogenic variants in other breast cancer susceptibility genes, including ATM, CHEK2, PALB2, RAD51C, RAD51D and BARD1, as well as genes associated with breast cancer predisposition syndromes - TP53 (Li-Fraumeni syndrome), PTEN (Cowden syndrome), CDH1 (hereditary diffuse gastric cancer), STK11 (Peutz-Jeghers syndrome) and NF1 (neurofibromatosis type 1). Polygenic risk, the cumulative risk from carrying multiple low-penetrance breast cancer susceptibility alleles, is also a well-recognised contributor to risk. This review provides an overview of the established breast cancer susceptibility genes as well as breast cancer predisposition syndromes, highlights distinct genotype-phenotype correlations associated with germline mutation status and discusses molecular testing and therapeutic implications in the context of hereditary breast cancer.
Collapse
Affiliation(s)
- A Sokolova
- Sullivan and Nicolaides PathologyBrisbane
- Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbane
| | - K J Johnstone
- Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbane
- Pathology Queensland, The Royal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - A E McCart Reed
- Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbane
| | - P T Simpson
- Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbane
| | - S R Lakhani
- Centre for Clinical Research, Faculty of MedicineThe University of QueenslandBrisbane
- Pathology Queensland, The Royal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| |
Collapse
|
163
|
Tsang JY, Tse GM. Update on triple-negative breast cancers - highlighting subtyping update and treatment implication. Histopathology 2023; 82:17-35. [PMID: 36468263 DOI: 10.1111/his.14784] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 12/12/2022]
Abstract
Triple-negative breast cancer (TNBC) remains a major challenge in breast cancer management. Continuing research in the past years aimed at understanding the biology of this tumour and developing more effective therapeutic options. It is now clear that TNBC is vastly heterogeneous with diverse histological, molecular, immunological profiles and clinical differences. Current evidence suggested the existence of at least four predominant subtypes based on expression profiling across studies. These subtypes exhibited specific genomic alterations and tumour microenvironment. Subtype-specific therapeutic strategies were identified. Recognising these subtypes allows not only an improved prognostication but also a better treatment decision. Herein, we provide an overview of the recent findings on TNBC heterogeneity at different levels and corresponding subtyping. The characteristic of subtypes and the implication of these subtypings in therapeutic approaches are also discussed.
Collapse
Affiliation(s)
- Julia Y Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
164
|
Herzog TJ, Vergote I, Gomella LG, Milenkova T, French T, Tonikian R, Poehlein C, Hussain M. Testing for homologous recombination repair or homologous recombination deficiency for poly (ADP-ribose) polymerase inhibitors: A current perspective. Eur J Cancer 2023; 179:136-146. [PMID: 36563604 DOI: 10.1016/j.ejca.2022.10.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022]
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPis) have demonstrated clinical activity in patients with BRCA1 and/or BRCA2 mutated breast, ovarian, prostate, and pancreatic cancers. Notably, BRCA mutations are associated with defects in the homologous recombination repair (HRR) pathway. This homologous recombination deficiency (HRD) phenotype can also be observed as genomic instability in tumour cells. Accordingly, PARPi sensitivity has been observed in various tumours with HRD, independent of BRCA mutations. Currently, four PARPis are approved by regulatory agencies for the treatment of cancer across multiple tumour types. Most indications are specific to tumours with a confirmed BRCA mutation, mutations in other HRR-related genes, HRD evidenced by genomic instability, or evidence of platinum sensitivity. Regulatory agencies have also approved companion and complementary diagnostics to facilitate patient selection for each PARPi indication. This review aims to summarise the biological basis, clinical validation, and clinical relevance of the available diagnostic methods and assays to assess HRD.
Collapse
Affiliation(s)
- Thomas J Herzog
- University of Cincinnati Cancer Center, University of Cincinnati Medical Center, 234 Goodman St, Cincinnati, OH 45219, USA.
| | - Ignace Vergote
- Department of Gynecology and Obstetrics, Division of Gynecologic Oncology, Leuven Cancer Institute, Catholic University Leuven, Herestraat 49, 3000 Leuven, Belgium, European Union
| | - Leonard G Gomella
- Department of Urology, Sidney Kimmel Cancer Center-Jefferson University Health, 1025 Walnut St Suite 1100, Philadelphia, PA 19107, USA
| | | | - Tim French
- AstraZeneca, 316 Hills Rd, Cambridge CB2 8PA, UK
| | - Raffi Tonikian
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ 07065, USA
| | | | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 420 E Superior St, Chicago, IL 60611, USA
| |
Collapse
|
165
|
Lazard T, Bataillon G, Naylor P, Popova T, Bidard FC, Stoppa-Lyonnet D, Stern MH, Decencière E, Walter T, Vincent-Salomon A. Deep learning identifies morphological patterns of homologous recombination deficiency in luminal breast cancers from whole slide images. Cell Rep Med 2022; 3:100872. [PMID: 36516847 PMCID: PMC9798078 DOI: 10.1016/j.xcrm.2022.100872] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/04/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
Homologous recombination DNA-repair deficiency (HRD) is becoming a well-recognized marker of platinum salt and polyADP-ribose polymerase inhibitor chemotherapies in ovarian and breast cancers. While large-scale screening for HRD using genomic markers is logistically and economically challenging, stained tissue slides are routinely acquired in clinical practice. With the objectives of providing a robust deep-learning method for HRD prediction from tissue slides and identifying related morphological phenotypes, we first show that digital pathology workflows are sensitive to potential biases in the training set, then we propose a method to overcome the influence of these biases, and we develop an interpretation method capable of identifying complex phenotypes. Application to our carefully curated in-house dataset allows us to predict HRD with high accuracy (area under the receiver-operator characteristics curve 0.86) and to identify morphological phenotypes related to HRD. In particular, the presence of laminated fibrosis and clear tumor cells associated with HRD open new hypotheses regarding its phenotypic impact.
Collapse
Affiliation(s)
- Tristan Lazard
- Center for Computational Biology (CBIO), Mines Paris, PSL University, 60 Boulevard Saint Michel, 75006 Paris, France,Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France
| | - Guillaume Bataillon
- Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France,Diagnostic and Theranostic Medicine Division, Institut Curie, PSL University, Paris, France
| | - Peter Naylor
- Center for Computational Biology (CBIO), Mines Paris, PSL University, 60 Boulevard Saint Michel, 75006 Paris, France,Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France
| | - Tatiana Popova
- INSERM U830, DNA Repair and Uveal Melanoma (DRUM), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, 75005 Paris, France
| | - François-Clément Bidard
- Department of Medical Oncology, Institut Curie, Université de Versailles Saint-Quentin, Saint-Cloud, France,INSERM CIC-BT 1428, Institut Curie, Paris, France
| | - Dominique Stoppa-Lyonnet
- INSERM U830, DNA Repair and Uveal Melanoma (DRUM), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, 75005 Paris, France,Université Paris Cité, 75006 Paris, France
| | - Marc-Henri Stern
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL University, Paris, France,INSERM U830, DNA Repair and Uveal Melanoma (DRUM), Equipe Labellisée par la Ligue Nationale Contre le Cancer, Institut Curie, PSL Research University, 75005 Paris, France
| | - Etienne Decencière
- Center for Mathematical Morphology (CMM), Mines Paris, PSL University, 77300 Fontainebleau, France
| | - Thomas Walter
- Center for Computational Biology (CBIO), Mines Paris, PSL University, 60 Boulevard Saint Michel, 75006 Paris, France,Institut Curie, PSL University, 75005 Paris, France,INSERM U900, 75005 Paris, France,Corresponding author
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL University, Paris, France,INSERM U934, CNRS UMR 3215, Paris, France,Corresponding author
| |
Collapse
|
166
|
Repurposing of Commercially Existing Molecular Target Therapies to Boost the Clinical Efficacy of Immune Checkpoint Blockade. Cancers (Basel) 2022; 14:cancers14246150. [PMID: 36551637 PMCID: PMC9776741 DOI: 10.3390/cancers14246150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/29/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Immune checkpoint blockade (ICB) is now standard of care for several metastatic epithelial cancers and prolongs life expectancy for a significant fraction of patients. A hostile tumor microenvironment (TME) induced by intrinsic oncogenic signaling induces an immunosuppressive niche that protects the tumor cells, limiting the durability and efficacy of ICB therapies. Addition of receptor tyrosine kinase inhibitors (RTKi) as potential modulators of an unfavorable local immune environment has resulted in moderate life expectancy improvement. Though the combination strategy of ICB and RTKi has shown significantly better results compared to individual treatment, the benefits and adverse events are additive whereas synergy of benefit would be preferable. There is therefore a need to investigate the potential of inhibitors other than RTKs to reduce malignant cell survival while enhancing anti-tumor immunity. In the last five years, preclinical studies have focused on using small molecule inhibitors targeting cell cycle and DNA damage regulators such as CDK4/6, CHK1 and poly ADP ribosyl polymerase (PARP) to selectively kill tumor cells and enhance cytotoxic immune responses. This review provides a comprehensive overview of the available drugs that attenuate immunosuppression and overcome hostile TME that could be used to boost FDA-approved ICB efficacy in the near future.
Collapse
|
167
|
Biomarkers and translational research approaches in breast cancer—an update. MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2022. [DOI: 10.1007/s12254-022-00855-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SummaryDiagnosis and decision-making in the treatment of breast cancer patients is vastly dependent on the exploration of biomarkers. Estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are long-standing biomarkers, which determine the breast cancer subtype. In current practice, gene expression analyses further define the molecular breast cancer subtype and give additional information on disease characteristics. Prognostic biomarkers provide information regarding recurrence risk and survival. Predictive biomarkers, such as programmed cell death ligand 1 expression, are tools for identifying patients who can benefit from specific therapy regimens in order to choose the best treatment option for the patient. While some biomarkers are affordable and readily available, others remain technically complex to access. Translational research builds the bridge from discovering novel biomarkers in preclinical studies to testing their application utility in the clinical setting. Integrating translational studies into clinical trials is therefore essential to find novel and reliable biomarkers for an optimal personalized treatment approach for patients with breast cancer.
Collapse
|
168
|
Clark CA, Yang ES. Therapeutic Targeting of DNA Damage Repair in the Era of Precision Oncology and Immune Checkpoint Inhibitors. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2022; 6:31-49. [PMID: 36751656 PMCID: PMC9888518 DOI: 10.36401/jipo-22-15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/08/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022]
Abstract
Cancer manifestation is a multistep process involving accumulation of various genetic and epigenetic changes that results in oncogenic "hallmarks of cancer" processes including genomic instability. Exploitation of aberrant DNA-damage response (DDR) mechanisms in cancer is in part a goal of many therapeutic strategies, and recent evidence supports the role of targeting DDR in modulating the tumor immune microenvironment to enhance immunotherapeutic response. Improved cancer profiling, including next-generation and whole-genome mutational sequencing of tumor tissue, as well as circulating nucleic acids, has enhanced our understanding of the genetic and epigenetic molecular mechanisms in tumorigenesis and will become fundamental to precisely target tumors and achieve cancer control. With the successes of poly(ADP-ribose) polymerase inhibitors (PARPi) and immunotherapies, the intersection of DDR molecular machinery and corresponding antitumor immune response has gained much interest with a focus on achieving therapeutic synergy using DNA damage-targeting agents and immunotherapy. In this review, we provide a bench-to-bedside overview of the fundamentals of DDR signaling and repair as they relate to cancer therapeutic strategies including novel DDR-targeting agents. We also discuss the underlying mechanisms that link DDR signaling to antitumor immunity and immunotherapy efficacy, and how this knowledge can be used to improve precision medicine approaches in the treatment of cancer.
Collapse
Affiliation(s)
- Curtis A. Clark
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Eddy S. Yang
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
,Hugh Kaul Precision Medicine Institute, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| |
Collapse
|
169
|
He R, Yuan X, Chen Z, Zheng Y. Combined immunotherapy for metastatic triple-negative breast cancer based on PD-1/PD-L1 immune checkpoint blocking. Int Immunopharmacol 2022; 113:109444. [DOI: 10.1016/j.intimp.2022.109444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
|
170
|
Miglietta F, Cinquini M, Dieci MV, Cortesi L, Criscitiello C, Montemurro F, Del Mastro L, Zambelli A, Biganzoli L, Levaggi A, Delle Piane C, Marchiò C, Calabrese M, Fortunato L, Franco P, Meduri B, Fittipaldo VA, Gori S. PARP-inhibitors for BRCA1/2-related advanced HER2-negative breast cancer: A meta-analysis and GRADE recommendations by the Italian Association of Medical Oncology. Breast 2022; 66:293-304. [PMID: 36379199 PMCID: PMC9663524 DOI: 10.1016/j.breast.2022.10.014] [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: 09/15/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Approximately 5-10% of unselected breast cancer (BC) patients retain a hereditary predisposition related to a germline mutation in BRCA1/2 genes. The poly-ADP ribose polymerase (PARP)-inhibitors olaparib and talazoparib have been granted marketing authorization by both FDA and EMA for adults with BRCA1/2 germline mutations and HER2-negative (HER2-) advanced BC based on the results from the phase III OlympiAd and EMBRACA trials. METHODS The panel of the Italian Association of Medical Oncology (AIOM) Clinical Practice Guidelines on Breast Cancer addressed two critical clinical questions, adopting the Grades of Recommendation, Assessment, Development and Evaluation (GRADE) approach and the Evidence to Decision framework (EtD), to develop recommendations on the use of PARP-inhibitors, with respect to single-agent chemotherapy, in patients with BRCA-related triple-negative (clinical question 1) and hormone receptor-positive (HR+)/HER2- (clinical question 2) advanced BC. RESULTS Two studies were eligible (OlympiAd and EMBRACA). For both clinical questions, the Panel judged the benefit/harm balance probably in favor of the intervention, given the favorable impact in terms of PFS, ORR, and QoL at an acceptable cost in terms of toxicity; the overall certainty of the evidence was low. The panel's final recommendations were conditional in favor of PARP-inhibitors over single-agent chemotherapy in both HR+/HER2-and triple-negative BC. Finally, the Panel identified and discussed areas of uncertainty calling for further exploration. CONCLUSIONS The Panel of AIOM BC Clinical Practice Guideline provided clinical recommendations on the use of PARP-inhibitors, with respect to single-agent chemotherapy, in patients with BRCA-related HER2-advanced BC by adopting the GRADE methodology.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy.
| | - Michela Cinquini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS., Laboratory of Methodology of Sistematic Reviews and Guidelines production; Department of Oncology, Milano, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Division of Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Laura Cortesi
- Department of Oncology and Hematology Azienda Ospedaliero-Universitaria di Modena, Italy
| | - Carmen Criscitiello
- European Institute of Oncology IRCCS, Milan, Italy; University of Milan, Milan, Italy
| | - Filippo Montemurro
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Lucia Del Mastro
- Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Internal Medicine and Medical Specialities, School of Medicine, University of Genova, Genova, Italy
| | - Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy; IRCCS Humanitas Research Hospital, Milan, Italy
| | | | - Alessia Levaggi
- Department of Oncology, Sant'Andrea Hospital, La Spezia, Italy
| | - Chiara Delle Piane
- Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Turin, Turin, Italy
| | - Massimo Calabrese
- Senologia Diagnostica, IRCCS-Ospedale Policlinico San Martino - Genova, Italy
| | - Lucio Fortunato
- Centro di Senologia - Azienda Ospedaliera San Giovanni-Addolorata, Roma, Italy
| | - Pierfrancesco Franco
- Department of Translational Medicine (DIMET), University of Eastern Piedmont, Novara, Italy
| | - Bruno Meduri
- Radioterapia - Azienda ospedaliera-Universitaria di Modena, Modena, Italy
| | - Veronica Andrea Fittipaldo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS., Laboratory of Methodology of Sistematic Reviews and Guidelines production; Department of Oncology, Milano, Italy
| | - Stefania Gori
- Medical Oncology Unit, Sacro Cuore - Don Calabria Hospital, Cancer Care Center, Negrar, Italy
| |
Collapse
|
171
|
O'Malley DM, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, Colombo N, Weberpals JI, Clamp AR, Scambia G, Leary A, Holloway RW, Gancedo MA, Fong PC, Goh JC, Swisher EM, Maloney L, Goble S, Lin KK, Kwan T, Ledermann JA, Coleman RL. Clinical and molecular characteristics of ARIEL3 patients who derived exceptional benefit from rucaparib maintenance treatment for high-grade ovarian carcinoma. Gynecol Oncol 2022; 167:404-413. [PMID: 36273926 PMCID: PMC10339359 DOI: 10.1016/j.ygyno.2022.08.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE ARIEL3 (NCT01968213) is a placebo-controlled randomized trial of the poly(ADP-ribose) polymerase inhibitor rucaparib as maintenance treatment in patients with recurrent high-grade ovarian carcinoma who responded to their latest line of platinum therapy. Rucaparib improved progression-free survival across all predefined subgroups. Here, we present an exploratory analysis of clinical and molecular characteristics associated with exceptional benefit from rucaparib. METHODS Patients were randomized 2:1 to receive rucaparib 600 mg twice daily or placebo. Molecular features (genomic alterations, BRCA1 promoter methylation) and baseline clinical characteristics were evaluated for association with exceptional benefit (progression-free survival ≥2 years) versus progression on first scan (short-term subgroup) and other efficacy outcomes. RESULTS Rucaparib treatment was significantly associated with exceptional benefit compared with placebo: 79/375 (21.1%) vs 4/189 (2.1%), respectively (p < 0.0001). Exceptional benefit was more frequent among patients with favorable baseline clinical characteristics and with carcinomas harboring molecular evidence of homologous recombination deficiency (HRD). A comparison between patients who derived exceptional benefit from rucaparib and those in the short-term subgroup revealed both clinical markers (no measurable disease at baseline, complete response to latest platinum, longer penultimate platinum-free interval) and molecular markers (BRCA1, BRCA2, RAD51C, and RAD51D alterations and genome-wide loss of heterozygosity) significantly associated with exceptional benefit. CONCLUSIONS Exceptional benefit in ARIEL3 was more common in, but not exclusive to, patients with favorable clinical characteristics or molecular features associated with HRD. Our results suggest that rucaparib can deliver exceptional benefit to a diverse set of patients with recurrent high-grade ovarian carcinoma.
Collapse
Affiliation(s)
- David M O'Malley
- Division of Gynecologic Oncology, The Ohio State University, James Cancer Center, Columbus, OH, USA. David.O'
| | - Amit M Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ana Oaknin
- Gynecologic Cancer Program, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Andrew Dean
- Department of Medical Oncology, St John of God Subiaco Hospital, Subaico, WA, Australia
| | - Nicoletta Colombo
- Department of Gynecologic Oncology, University of Milan-Bicocca and European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Johanne I Weberpals
- Department of Obstetrics and Gynecology, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andrew R Clamp
- Medical Oncology, The Christie NHS Foundation Trust and University of Manchester, Manchester, UK
| | - Giovanni Scambia
- Department of Cancer Gynecology, Fondazione Policlinico Universitario A. Gemelli IRCCS and Scientific Directorate, Rome, Italy
| | - Alexandra Leary
- Gynecological Unit, Gustave Roussy Cancer Center, INSERM U981, Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Villejuif, France
| | - Robert W Holloway
- Gynecologic Oncology, Florida Hospital Cancer Institute, Orlando, FL, USA
| | | | - Peter C Fong
- Medical Oncology, Auckland City Hospital and University of Auckland, New Zealand
| | - Jeffrey C Goh
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Australia; Faculty of Medicine, University of Queensland, St Lucia, Australia
| | - Elizabeth M Swisher
- Division of Gynecologic Oncology, University of Washington, Seattle, WA, USA
| | - Lara Maloney
- Clinical Development, Clovis Oncology, Inc., Boulder, CO, USA
| | - Sandra Goble
- Biostatistics, Clovis Oncology, Inc., Boulder, CO, USA
| | - Kevin K Lin
- Molecular Diagnostics, Clovis Oncology, Inc., Boulder, CO, USA
| | - Tanya Kwan
- Molecular Diagnostics, Clovis Oncology, Inc., Boulder, CO, USA
| | - Jonathan A Ledermann
- Department of Oncology, UCL Cancer Institute, University College London and UCL Hospitals, London, UK
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
172
|
The BRCAness Landscape of Cancer. Cells 2022; 11:cells11233877. [PMID: 36497135 PMCID: PMC9738094 DOI: 10.3390/cells11233877] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/23/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
BRCAness refers to the damaged homologous recombination (HR) function due to the defects in HR-involved non-BRCA1/2 genes. BRCAness is the important marker for the use of synthetic lethal-based PARP inhibitor therapy in breast and ovarian cancer treatment. The success provides an opportunity of applying PARP inhibitor therapy to treat other cancer types with BRCAness features. However, systematic knowledge is lack for BRCAness in different cancer types beyond breast and ovarian cancer. We performed a comprehensive characterization for 40 BRCAness-related genes in 33 cancer types with over 10,000 cancer cases, including pathogenic variation, homozygotic deletion, promoter hypermethylation, gene expression, and clinical correlation of BRCAness in each cancer type. Using BRCA1/BRCA2 mutated breast and ovarian cancer as the control, we observed that BRCAness is widely present in multiple cancer types. Based on the sum of the BRCAneass features in each cancer type, we identified the following 21 cancer types as the potential targets for PARPi therapy: adrenocortical carcinoma, bladder urothelial carcinoma, brain lower grade glioma, colon adenocarcinoma, esophageal carcinoma, head and neck squamous carcinoma, kidney chromophobe, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, rectum adenocarcinoma, pancreatic adenocarcinoma, prostate adenocarcinoma, sarcoma, skin cutaneous melanoma, stomach adenocarcinoma, uterine carcinosarcoma, and uterine corpus endometrial carcinoma.
Collapse
|
173
|
Oshi M, Gandhi S, Wu R, Asaoka M, Yan L, Yamada A, Yamamoto S, Narui K, Chishima T, Ishikawa T, Endo I, Takabe K. Development of a novel BRCAness score that predicts response to PARP inhibitors. Biomark Res 2022; 10:80. [PMID: 36371386 PMCID: PMC9652967 DOI: 10.1186/s40364-022-00427-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/30/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND BRCAness is a characteristic feature of homologous recombination deficiency (HRD) mimicking BRCA gene mutation in breast cancer. We hypothesized that a measure to quantify BRCAness that causes synthetic lethality in BRCA mutated tumors will identify responders to PARP inhibitors. METHODS A total of 6753 breast cancer patients from 3 large independent cohorts were analyzed. A score was generated by transcriptomic profiling using gene set variation analysis algorithm on 34 BRCA1-mutation related genes selected by high AUC levels in ROC curve between BRCA1 mutation and wildtype breast cancer. RESULTS The score was significantly associated with BRCA1 mutation, high mutation load and intratumoral heterogeneity as expected, as well as with high HRD, DNA repair and MKi67 expression regardless of BRCA mutations. High BRCAness tumors enriched not only DNA repair, but also all five Hallmark cell proliferation-related gene sets. High BRCAness tumors were significantly associated with higher cytolytic activity and with higher anti-cancerous immune cell infiltration. Not only did the breast cancer cell lines with BRCA-mutation show high score, but even the other cells in human breast cancer tumor microenvironment were contributing to the score. The BRCAness score was the highest in triple-negative breast cancer consistently in all 3 cohorts. BRCAness was associated with response to chemotherapy and correlated strongly with response to PARP inhibitor in both triple-negative and ER-positive/HER2-negative breast cancer. CONCLUSIONS We established a novel BRCAness score using BRCA-mutation-related gene expressions and found that it associates with DNA repair and predicts response to PARP inhibitors regardless of BRCA mutation.
Collapse
Affiliation(s)
- Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004 Japan
| | - Shipra Gandhi
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
| | - Rongrong Wu
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402 Japan
| | - Mariko Asaoka
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402 Japan
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
| | - Akimitsu Yamada
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004 Japan
| | - Shinya Yamamoto
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Kanagawa Japan
| | - Kazutaka Narui
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Kanagawa Japan
| | - Takashi Chishima
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004 Japan
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, 160-8402 Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004 Japan
| | - Kazuaki Takabe
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263 USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004 Japan
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8520 Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, 960-1295 Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14263 USA
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263 USA
| |
Collapse
|
174
|
Sibilio A, Curcio A, Toesca A, Rossi EMC, Corso G. Local treatment in patients with hereditary breast cancer: decision-making process in low-, moderate-, high-penetrance pathogenic germline mutation carriers. Curr Opin Oncol 2022; 34:614-622. [PMID: 36170166 DOI: 10.1097/cco.0000000000000872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW We summarize recent evidence regarding commonly tested breast cancer susceptibility genes and review indications derived from recently published guidelines regarding management of carriers affected by early breast cancer (BC). RECENT FINDINGS Management of affected women with a known genetic predisposition to BC was matter of debate at the most relevant international conferences, such as St. Gallen International Consensus Conference and San Antonio Breast Cancer Symposium held both in 2021. At the same time, a joint Experts Panel from American Society of Clinical Oncology/American Society for Radiation Oncology/Society of Surgical Oncology (ASCO/ASTRO/SSO) convened to develop recommendations to support clinical decision-making in this specific setting and results about administration of new systemic therapies such as poly adenosine diphosphate-ribose polymerase (PARP) inhibitors became available. SUMMARY Population of patients affected by BC and carriers of mutations in susceptibility genes is progressively increasing, but new mutations identified do not always have a clear clinical impact.To date, we have data to support consideration of different local management choices for affected patients carrying specific mutations, but some issues especially relating to breast-conserving surgery or administration of radiotherapy in these patients, still need to be better addressed. Opinions about the best way to treat these patients are still heterogeneous and information deriving from different sources seems to be conflicting at times. Our purpose is to offer a synopsis of the different evidence available that may be helpful in clinical decision making.
Collapse
Affiliation(s)
- Andrea Sibilio
- UOC Chirurgia Senologica Forlì-Ravenna, AUSL Romagna, Forlì, Italy
| | - Annalisa Curcio
- UOC Chirurgia Senologica Forlì-Ravenna, AUSL Romagna, Forlì, Italy
| | - Antonio Toesca
- Division of Breast Surgery, Candiolo Cancer Institute, IRCCS, Candiolo, Italy
| | | | - Giovanni Corso
- Division of Breast Surgery, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- European Cancer Prevention Organization (ECP), Milan, Italy
| |
Collapse
|
175
|
Taurelli Salimbeni B, Corvaja C, Valenza C, Zagami P, Curigliano G. The triple negative breast cancer drugs graveyard: a review of failed clinical trials 2017-2022. Expert Opin Investig Drugs 2022; 31:1203-1226. [PMID: 36413823 DOI: 10.1080/13543784.2022.2151433] [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] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) accounts for 15-20% of breast cancers (BC) and has the worst prognosis. It is characterized by the absence of both hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2). TNBC has more limited therapeutic options compared to other subtypes, meaning that there is still a long way to go to discover target treatments. AREAS COVERED Our review aims to summarize phase II/III clinical trials enrolling patients with TNBC that have been published between 2017 and 2022 but failed to reach their primary endpoint. We here try to emphasize the limitations and weaknesses noted in negative studies and to point out unexpected results which might be useful to enhance the therapeutic approach to TNBC disease. EXPERT OPINION A deeper understanding of the mechanisms behind TNBC heterogeneity allowed to enhance the knowledge of new prognostic and predictive biomarkers of response. However, it is also through several failed clinical trials that we were able to define new therapeutic approaches which improved TNBC patients' clinical outcomes. Nowadays, we still need to overcome several difficulties to fully recognize different intracellular and extracellular pathways that crosstalk in TNBC and the mechanisms of resistance to identify novel tailored-patients' therapies.
Collapse
Affiliation(s)
- Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Clinical and Molecular Medicine, Oncology Unit, "la Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - Carla Corvaja
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Medicine, University of Udine, Udine, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| | - Paola Zagami
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Irccs, Milan, Italy.,Department of Oncology and Haematology, University of Milan, Milan, Italy
| |
Collapse
|
176
|
McDonald JT, Ricks-Santi LJ. Hereditary variants of unknown significance in African American women with breast cancer. PLoS One 2022; 17:e0273835. [PMID: 36315513 PMCID: PMC9621418 DOI: 10.1371/journal.pone.0273835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/17/2022] [Indexed: 11/06/2022] Open
Abstract
Expanded implementation of genetic sequencing has precipitously increased the discovery of germline and somatic variants. The direct benefit of identifying variants in actionable genes may lead to risk reduction strategies such as increased surveillance, prophylactic surgery, as well as lifestyle modifications to reduce morbidity and mortality. However, patients with African ancestry are more likely to receive inconclusive genetic testing results due to an increased number of variants of unknown significance decreasing the utility and impact on disease management and prevention. This study examines whole exome sequencing results from germline DNA samples in African American women with a family history of cancer including 37 cases that were diagnosed with breast cancer and 51 family members. Self-identified ancestry was validated and compared to the 1000 genomes population. The analysis of sequencing results was limited to 85 genes from three clinically available common genetic screening platforms. This target region had a total of 993 variants of which 6 (<1%) were pathogenic or likely pathogenic, 736 (74.1%) were benign, and 170 (17.1%) were classified as a variant of unknown significance. There was an average of 3.4±1.8 variants with an unknown significance per individual and 85 of 88 individuals (96.6%) harbored at least one of these in the targeted genes. Pathogenic or likely pathogenic variants were only found in 6 individuals for the BRCA1 (p.R1726fs, rs80357867), BRCA2 (p.K589fs, rs397507606 & p.L2805fs, rs397507402), RAD50 (p.E995fs, rs587780154), ATM (p.V2424G, rs28904921), or MUTYH (p.G396D, rs36053993) genes. Strategies to functionally validate the remaining variants of unknown significance, especially in understudied and hereditary cancer populations, are greatly needed to increase the clinical utility and utilization of clinical genetic screening platforms to reduce cancer incidence and mortality.
Collapse
Affiliation(s)
- J. Tyson McDonald
- Department of Radiation Medicine, Georgetown University School of Medicine, Washington, DC, United States of America
| | - Luisel J. Ricks-Santi
- Cancer Research Center, Hampton University, Hampton, VA, United States of America
- Department of Pharmacotherapy and Translational Research, College of Medicine, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
177
|
Li S, Wang L, Wang Y, Zhang C, Hong Z, Han Z. The synthetic lethality of targeting cell cycle checkpoints and PARPs in cancer treatment. J Hematol Oncol 2022; 15:147. [PMID: 36253861 PMCID: PMC9578258 DOI: 10.1186/s13045-022-01360-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Continuous cell division is a hallmark of cancer, and the underlying mechanism is tumor genomics instability. Cell cycle checkpoints are critical for enabling an orderly cell cycle and maintaining genome stability during cell division. Based on their distinct functions in cell cycle control, cell cycle checkpoints are classified into two groups: DNA damage checkpoints and DNA replication stress checkpoints. The DNA damage checkpoints (ATM-CHK2-p53) primarily monitor genetic errors and arrest cell cycle progression to facilitate DNA repair. Unfortunately, genes involved in DNA damage checkpoints are frequently mutated in human malignancies. In contrast, genes associated with DNA replication stress checkpoints (ATR-CHK1-WEE1) are rarely mutated in tumors, and cancer cells are highly dependent on these genes to prevent replication catastrophe and secure genome integrity. At present, poly (ADP-ribose) polymerase inhibitors (PARPi) operate through “synthetic lethality” mechanism with mutant DNA repair pathways genes in cancer cells. However, an increasing number of patients are acquiring PARP inhibitor resistance after prolonged treatment. Recent work suggests that a combination therapy of targeting cell cycle checkpoints and PARPs act synergistically to increase the number of DNA errors, compromise the DNA repair machinery, and disrupt the cell cycle, thereby increasing the death rate of cancer cells with DNA repair deficiency or PARP inhibitor resistance. We highlight a combinational strategy involving PARP inhibitors and inhibition of two major cell cycle checkpoint pathways, ATM-CHK2-TP53 and ATR-CHK1-WEE1. The biological functions, resistance mechanisms against PARP inhibitors, advances in preclinical research, and clinical trials are also reviewed.
Collapse
Affiliation(s)
- Shuangying Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Liangliang Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yuanyuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Changyi Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
178
|
Graffeo R, Rana H, Conforti F, Bonanni B, Cardoso M, Paluch-Shimon S, Pagani O, Goldhirsch A, Partridge A, Lambertini M, Garber J. Moderate penetrance genes complicate genetic testing for breast cancer diagnosis: ATM, CHEK2, BARD1 and RAD51D. Breast 2022; 65:32-40. [PMID: 35772246 PMCID: PMC9253488 DOI: 10.1016/j.breast.2022.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer risk associated with germline likely pathogenic/pathogenic variants (PV) varies by gene, often by penetrance (high >50% or moderate 20–50%), and specific locus. Germline PVs in BRCA1 and BRCA2 play important roles in the development of breast and ovarian cancer in particular, as well as in other cancers such as pancreatic and prostate cancers and melanoma. Recent studies suggest that other cancer susceptibility genes, including ATM, CHEK2, PALB2, RAD51C and RAD51D confer differential risks of breast and other specific cancers. In the era of multigene panel testing, advances in next-generation sequencing technologies have notably reduced costs in the United States (US) and enabled sequencing of BRCA1/2 concomitantly with additional genes. The use of multigene-panel testing is beginning to expand in Europe as well. Further research into the clinical implications of variants in moderate penetrance genes, particularly in unaffected carriers, is needed for appropriate counselling and risk management with data-driven plans for surveillance and/or risk reduction. For individuals at high risk without any pathogenic or likely pathogenic variant in cancer susceptibility genes or some carriers of pathogenic variants in moderate-risk genes such as ATM and CHEK2, polygenic risk scores offer promise to help stratify breast cancer risk and guide appropriate risk management options. Cancer patients whose tumours are driven by the loss of function of both copies of a predisposition gene may benefit from therapies targeting the biological alterations induced by the dysfunctional gene e.g. poly ADP ribose polymerase (PARP) inhibitors and other novel pathway agents in cancers with DNA repair deficiencies. A better understanding of mechanisms by which germline variants drive various malignancies may lead to improvements in both therapeutic and preventive management options.
The interpretation of genetic testing results requires careful attention. ATM, CHEK2, RAD51D and BARD1 correlated with breast and other cancers risk. European and American guidelines discrepancies. Support European healthcare providers in interpreting and managing female carriers.
Collapse
|
179
|
Affiliation(s)
- Florence Coussy
- Institut Curie, Department of Medical Oncology, Paris and Saint-Cloud, France
| | - Francois-Clement Bidard
- Institut Curie, Department of Medical Oncology, Paris and Saint-Cloud, France.
- Université Paris-Saclay, UVSQ, Saint-Cloud, France.
- Inserm CIC-BT 1428, Paris, France.
| |
Collapse
|
180
|
Andre F, Filleron T, Kamal M, Mosele F, Arnedos M, Dalenc F, Sablin MP, Campone M, Bonnefoi H, Lefeuvre-Plesse C, Jacot W, Coussy F, Ferrero JM, Emile G, Mouret-Reynier MA, Thery JC, Isambert N, Mege A, Barthelemy P, You B, Hajjaji N, Lacroix L, Rouleau E, Tran-Dien A, Boyault S, Attignon V, Gestraud P, Servant N, Le Tourneau C, Cherif LL, Soubeyran I, Montemurro F, Morel A, Lusque A, Jimenez M, Jacquet A, Gonçalves A, Bachelot T, Bieche I. Genomics to select treatment for patients with metastatic breast cancer. Nature 2022; 610:343-348. [PMID: 36071165 DOI: 10.1038/s41586-022-05068-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 07/03/2022] [Indexed: 01/04/2023]
Abstract
Cancer progression is driven in part by genomic alterations1. The genomic characterization of cancers has shown interpatient heterogeneity regarding driver alterations2, leading to the concept that generation of genomic profiling in patients with cancer could allow the selection of effective therapies3,4. Although DNA sequencing has been implemented in practice, it remains unclear how to use its results. A total of 1,462 patients with HER2-non-overexpressing metastatic breast cancer were enroled to receive genomic profiling in the SAFIR02-BREAST trial. Two hundred and thirty-eight of these patients were randomized in two trials (nos. NCT02299999 and NCT03386162) comparing the efficacy of maintenance treatment5 with a targeted therapy matched to genomic alteration. Targeted therapies matched to genomics improves progression-free survival when genomic alterations are classified as level I/II according to the ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT)6 (adjusted hazards ratio (HR): 0.41, 90% confidence interval (CI): 0.27-0.61, P < 0.001), but not when alterations are unselected using ESCAT (adjusted HR: 0.77, 95% CI: 0.56-1.06, P = 0.109). No improvement in progression-free survival was observed in the targeted therapies arm (unadjusted HR: 1.15, 95% CI: 0.76-1.75) for patients presenting with ESCAT alteration beyond level I/II. Patients with germline BRCA1/2 mutations (n = 49) derived high benefit from olaparib (gBRCA1: HR = 0.36, 90% CI: 0.14-0.89; gBRCA2: HR = 0.37, 90% CI: 0.17-0.78). This trial provides evidence that the treatment decision led by genomics should be driven by a framework of target actionability in patients with metastatic breast cancer.
Collapse
Affiliation(s)
- Fabrice Andre
- Department of Medical Oncology, Gustave Roussy, Villejuif, France. .,INSERM U981, Gustave Roussy, Villejuif, France. .,PRISM Center for personalized medicine, Gustave Roussy, Villejuif, France. .,Medical School, Université Paris Saclay, Kremlin Bicetre, France.
| | - Thomas Filleron
- Department of Biostatistics, Institut Claudius Regaud, IUCT oncopole, Toulouse, France
| | - Maud Kamal
- Department of Drug Development and Innovation, Institut Curie, Saint Cloud, France
| | | | - Monica Arnedos
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Florence Dalenc
- Department of Medical Oncology, Institut Claudius-Regaud IUCT oncopole and University of Paul Sabatier, Toulouse, France
| | - Marie-Paule Sablin
- Department of Drug Development and Innovation, Institut Curie, Saint Cloud, France.,Department of Medical Oncology, Institut Curie, Paris, France
| | - Mario Campone
- Institut de Cancérologie de l'Ouest - René Gauducheau, Saint Herblain, University of Angers, Angers, France
| | - Hervé Bonnefoi
- Department of Medical Oncology, Institut Bergonié INSERM U1218 and Université of Bordeaux, Bordeaux, France
| | | | - William Jacot
- Department of Medical Oncology, Institut du Cancer de Montpellier, Institut de Recherche en Cancérologie de Montpellier INSERM U1194 and Montpellier University, Montpellier, France
| | - Florence Coussy
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France
| | - Jean-Marc Ferrero
- Department of Medical Oncology, Centre Antoine Lacassagne, University Côte d'Azur, Nice, France
| | - George Emile
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | | | - Jean-Christophe Thery
- Department of Medical Oncology, Centre Hennri Becquerel, University of Medicine of Rouen, Rouen, France
| | - Nicolas Isambert
- Department of Medical Oncology, Centre Georges François Leclerc, Dijon, France
| | - Alice Mege
- Institut Sainte Catherine, Avignon, France
| | | | - Benoit You
- Department of Medical Oncology, Institut de Cancérologie des Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Nawale Hajjaji
- Department of Medical Oncology, Centre Oscar Lambret INSERM U1192 PRISM Laboratory and University of Lille, Lille, France
| | - Ludovic Lacroix
- Cancer Genetics Laboratory, Department of Pathology and Medical Biology, Gustave Roussy, Villejuif, France
| | - Etienne Rouleau
- Cancer Genetics Laboratory, Department of Pathology and Medical Biology, Gustave Roussy, Villejuif, France
| | - Alicia Tran-Dien
- INSERM U981, Gustave Roussy, Villejuif, France.,PRISM Center for personalized medicine, Gustave Roussy, Villejuif, France.,Bioinformatic Core Facility, UMS AMMICA, Gustave Roussy, Villejuif, France
| | - Sandrine Boyault
- Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Valery Attignon
- Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Pierre Gestraud
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Nicolas Servant
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | | | - Linda Larbi Cherif
- Department of Drug Development and Innovation, Institut Curie, Saint Cloud, France
| | - Isabelle Soubeyran
- Unit of Molecular Pathology - Department of Biopathology, Institut Bergonié, Bordeaux, France
| | | | - Alain Morel
- Department of Innate Immunity and Immunotherapy, Institut de Cancérologie de l'Ouest - Centre Paul Papin, Angers, France
| | - Amelie Lusque
- Department of Biostatistics, Institut Claudius Regaud, IUCT oncopole, Toulouse, France
| | | | | | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Ivan Bieche
- Department of Genetics, Institut Curie, INSERM U1016, Université Paris Cité, Paris, France
| |
Collapse
|
181
|
Untch M, Fasching PA, Haidinger R, Harbeck N, Jackisch C, Lüftner D, Müller V, Schumacher-Wulf E, Würstlein R, Thomssen C. Advanced Breast Cancer: AGO Recommendations 2022 - Focus on ABC6 Consensus. Geburtshilfe Frauenheilkd 2022; 82:1044-1054. [PMID: 36186150 PMCID: PMC9525147 DOI: 10.1055/a-1904-6100] [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: 05/15/2022] [Accepted: 07/18/2022] [Indexed: 12/05/2022] Open
Abstract
Over the past few years, there have been many developments in the treatment of advanced breast cancer; these have been incorporated into national and international treatment guidelines, resulting in an improved prognosis for these patients. The 6th International Consensus Conference for Advanced Breast Cancer (ABC6) was held in November 2021. The aim is to standardize the treatment of advanced breast cancer based on a high level of evidence, and to make new treatment options accessible to all patients. In this article we discuss the ABC6 consensus in the context of German treatment guidelines, and compare it with clinical practice in Germany. The authors refer to the current recommendations of the Breast Cancer Working Group for Gynecological Oncology (AGO Mamma) published in March 2022. The AGO Breast Cancer Guidelines are updated annually. Since discrepancies between national and international guidelines can occur due to country-specific regulations, this is a useful comparison to make. The German authors refer to the voting results of the ABC6 panelists from 6 November 2021.
Collapse
Affiliation(s)
- Michael Untch
- Klinik für Gynäkologie und Geburtshilfe, Multidisziplinäres Brustzentrum und Zentrum für gynäkologische Onkologie, HELIOS Klinikum Berlin Buch, Berlin, Germany
| | - Peter A. Fasching
- Universitätsfrauenklinik Erlangen, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renate Haidinger
- Brustkrebs Deutschland e. V., Hohenbrunn, Germany; Patientenvertreterin, ABC6-Ko-Vorsitz, ABC-Panelistin
| | - Nadia Harbeck
- Brustzentrum, Klinik und Poliklinik für Frauenheilkunde und Comprehensive Cancer Center (CCC) München, LMU Klinikum, München, Germany; ABC-Panelistin
| | - Christian Jackisch
- Klinik für Gynäkologie und Geburtshilfe, Sana Klinikum Offenbach GmbH, Offenbach, Germany
| | - Diana Lüftner
- Immanuel Klinik Märkische Schweiz, Medizinische Hochschule Brandenburg Theodor-Fontane, Buckow, Germany
| | - Volkmar Müller
- Universitätsklinik für Gynäkologie, Hamburg-Eppendorf, Hamburg, Germany
| | | | - Rachel Würstlein
- Brustzentrum, Klinik und Poliklinik für Frauenheilkunde und Comprehensive Cancer Center (CCC) München, LMU Klinikum, München, Germany
| | - Christoph Thomssen
- Universitätsfrauenklinik, Martin-Luther-Universität, Halle an der Saale, Germany; ABC-Panelist
| |
Collapse
|
182
|
Lai YH, Tung KC, Chen SC. Durable response to olaparib combined low-dose cisplatin in advanced hepatocellular carcinoma with FANCA mutation: A case report. Medicine (Baltimore) 2022; 101:e30719. [PMID: 36181052 PMCID: PMC9524966 DOI: 10.1097/md.0000000000030719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RATIONALE To date, there is no actionable gene has been discovered in hepatocellular carcinoma (HCC). Tumor cells with DNA damage response and repair (DDR) gene loss-of-function mutation is sensitivity to poly-ADP-ribose polymerase (PARP) inhibitors and platinum chemotherapy in ovarian, prostate and pancreatic cancers. There is a case report demonstrated the efficacy of PARP inhibitor for BRCA2 mutation that belongs to DDR gene in HCC, which suggested the potential role of PARP inhibitor for HCC with DDR gene mutation. PATIENT CONCERNS We reported a 44-year-old woman with non-viral HCC who was refractory to multiple treatment including target therapy, immunotherapy, and chemotherapy. The tumor tissue was submitted to next-generation sequencing using the commercially available ACTOnco®+ (ACT Genomics, Taiwan) assay that interrogates 440 and 31 cancer-related genes and fusion genes, respectively. DIAGNOSIS A truncating mutation FANCA p.Q1307fs was also observed. The tumor was microsatellite stable and had low tumor mutational burden of 4.5 muts/Mb. INTERVENTIONS AND OUTCOMES Given FANCA belongs to DDR genes, the inactivation evoked the idea of using PARP inhibitor and cisplatin. Therefore, the patient started to use olaparib combined with low-dose cisplatin (30 mg/m2, every 4 weeks) therapy in December 2019. Significant reduction in the tumor marker level in 1 month (PIVKA-II from 17,395 to 411 ng/dL) and follow-up CT scan showed stable disease. Her tumor did not progress until December 2020 with a progression-free survival of 12 months. LESSONS We report the first case of FANCA-mutated HCC that responded well to olaparib and low-dose cisplatin. This addressed the potential therapeutic role of DDR gene mutation in HCC and the possible synergistic effect of PARP inhibitor and cisplatin. These findings highlight areas where further investigation and effort are needed.
Collapse
Affiliation(s)
- Yi-Hsuan Lai
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kai-Che Tung
- ACT Genomics Co., Ltd, Taipei, Taiwan
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - San-Chi Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Medical Oncology, Center of Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- *Correspondence: San-Chi Chen, Division of Medical Oncology, Center for Immuno-oncology, Department of Oncology, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Road, Taipei 11217, Taiwan (e-mail: )
| |
Collapse
|
183
|
CDK14 inhibition reduces mammary stem cell activity and suppresses triple negative breast cancer progression. Cell Rep 2022; 40:111331. [PMID: 36103813 DOI: 10.1016/j.celrep.2022.111331] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/09/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays an important role in regulating mammary organogenesis and oncogenesis. However, therapeutic methods targeting the Wnt pathway against breast cancer have been limited. To address this challenge, we investigate the function of cyclin-dependent kinase 14 (CDK14), a member of the Wnt signaling pathway, in mammary development and breast cancer progression. We show that CDK14 is expressed in the mammary basal layer and elevated in triple negative breast cancer (TNBC). CDK14 knockdown reduces the colony-formation ability and regeneration capacity of mammary basal cells and inhibits the progression of murine MMTV-Wnt-1 basal-like mammary tumor. CDK14 knockdown or pharmacological inhibition by FMF-04-159-2 suppresses the progression and metastasis of TNBC. Mechanistically, CDK14 inhibition inhibits mammary regeneration and TNBC progression by attenuating Wnt/β-catenin signaling. These findings highlight the significance of CDK14 in mammary development and TNBC progression, shedding light on CDK14 as a promising therapeutic target for TNBC.
Collapse
|
184
|
Paul S, Sinha S, Kundu CN. Targeting cancer stem cells in the tumor microenvironment: An emerging role of PARP inhibitors. Pharmacol Res 2022; 184:106425. [PMID: 36075511 DOI: 10.1016/j.phrs.2022.106425] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022]
Abstract
Cancer stem cells (CSCs) constitute a small population of cancer cells in the tumor microenvironment (TME), which are responsible for metastasis, angiogenesis, drug resistance, and cancer relapse. Understanding the key signatures and resistance mechanisms of CSCs may help in the development of novel chemotherapeutic strategies to specifically target CSCs in the TME. PARP inhibitors (PARPi) are known to enhance the chemosensitivity of cancer cells to other chemotherapeutic agents by inhibiting the DNA repair pathways and chromatin modulation. But their effects on CSCs are still unknown. Few studies have reported that PARPi can stall replication fork progression in CSCs. PARPi also have the potential to overcome chemoresistance in CSCs and anti-angiogenic potentiality as well. Previous reports have suggested that epigenetic drugs can synergistically ameliorate the anti-cancer activities of PARPi through epigenetic modulations. In this review, we have systematically discussed the effects of PARPi on different DNA repair pathways with respect to CSCs and also how CSCs can be targeted either as monotherapy or as a part of combination therapy. We have also talked about how PARPi can help in reversal of chemoresistance of CSCs and the role of PARPi in epigenetic modifications to hinder cancer progression. We have also elaborated on the aspects of research that need to be investigated for development of successful therapeutic interventions using PARPi to specifically target CSCs in the TME.
Collapse
Affiliation(s)
- Subarno Paul
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Saptarshi Sinha
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
185
|
PARP Inhibitors for Breast Cancer: Germline BRCA1/2 and Beyond. Cancers (Basel) 2022; 14:cancers14174332. [PMID: 36077867 PMCID: PMC9454726 DOI: 10.3390/cancers14174332] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/27/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Poly-adenosine diphosphate ribose polymerase (PARP) inhibitors (PARPi) are effective against tumors with mutations in DNA repair genes, most commonly in the BRCA1 and BRCA2 genes. Because these tumors are unable to repair their DNA, PARPi have been used to target DNA repair pathways and are useful in the treatment of breast cancers with some of these alterations. There are two FDA-approved PARPi for patients with breast cancer—olaparib and talazoparib. The data on olaparib and talazoparib in the treatment of breast cancer are summarized in this review, and we also explore potential future applications of PARPi beyond inherited BRCA mutations. Abstract Poly-adenosine diphosphate ribose polymerase (PARP) inhibitors (PARPi) are approved for BRCA1/2 carriers with HER2-negative breast cancer in the adjuvant setting with a high risk of recurrence as well as the metastatic setting. However, the indications for PARPi are broader for patients with other cancer types (e.g., prostate and ovarian cancer), involving additional biomarkers (e.g., ATM, PALB2, and CHEK) and genomic instability scores. Herein, we summarize the data on PARPi and breast cancer and discuss their use beyond BRCA carriers.
Collapse
|
186
|
Heterogeneity of triple negative breast cancer: Current advances in subtyping and treatment implications. J Exp Clin Cancer Res 2022; 41:265. [PMID: 36050786 PMCID: PMC9434975 DOI: 10.1186/s13046-022-02476-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/25/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractAs the field of translational ‘omics has progressed, refined classifiers at both genomic and proteomic levels have emerged to decipher the heterogeneity of breast cancer in a clinically-applicable way. The integration of ‘omics knowledge at the DNA, RNA and protein levels is further expanding biologic understanding of breast cancer and opportunities for customized treatment, a particularly pressing need in clinically triple negative tumors. For this group of aggressive breast cancers, work from multiple groups has now validated at least four major biologically and clinically distinct omics-based subtypes. While to date most clinical trial designs have considered triple negative breast cancers as a single group, with an expanding arsenal of targeted therapies applicable to distinct biological pathways, survival benefits may be best realized by designing and analyzing clinical trials in the context of major molecular subtypes. While RNA-based classifiers are the most developed, proteomic classifiers proposed for triple negative breast cancer based on new technologies have the potential to more directly identify the most clinically-relevant biomarkers and therapeutic targets. Phospho-proteomic data further identify targetable signalling pathways in a unique subtype-specific manner. Single cell profiling of the tumor microenvironment represents a promising way to allow a better characterization of the heterogeneity of triple negative breast cancer which could be integrated in a spatially resolved context to build an ecosystem-based patient classification. Multi-omic data further allows in silico analysis of genetic and pharmacologic screens to map therapeutic vulnerabilities in a subtype-specific context. This review describes current knowledge about molecular subtyping of triple negative breast cancer, recent advances in omics-based genomics and proteomics diagnostics addressing the diversity of this disease, key advances made through single cell analysis approaches, and developments in treatments including targeted therapeutics being tested in major clinical trials.
Collapse
|
187
|
Schettini F, Venturini S, Giuliano M, Lambertini M, Pinato DJ, Elisa Onesti C, De Placido P, Harbeck N, Lüftner D, Denys H, Van Dam P, Arpino G, Zaman K, Mustacchi G, Gligorov J, Awada A, Campone M, Wildiers H, Gennari A, Tjan-Heijnen V, Bartsch R, Cortes J, Paris I, Martín M, De Placido S, Del Mastro L, Jerusalem G, Curigliano G, Prat A, Generali D. Multiple Bayesian Network Meta-Analyses to Establish Therapeutic Algorithms for Metastatic Triple Negative Breast Cancer. Cancer Treat Rev 2022; 111:102468. [DOI: 10.1016/j.ctrv.2022.102468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 12/23/2022]
|
188
|
Verret B, Bottosso M, Hervais S, Pistilli B. The Molecular Predictive and Prognostic Biomarkers in Metastatic Breast Cancer: The Contribution of Molecular Profiling. Cancers (Basel) 2022; 14:4203. [PMID: 36077738 PMCID: PMC9454488 DOI: 10.3390/cancers14174203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 08/20/2022] [Indexed: 11/22/2022] Open
Abstract
The past decade was marked by several important studies deciphering the molecular landscape of metastatic breast cancer. Although the initial goal of these studies was to find driver oncogenic events to explain cancer progression and metastatic spreading, they have also permitted the identification of several molecular alterations associated with treatment response or resistance. Herein, we review validated (PI3KCA, ESR1, MSI, NTRK translocation) and emergent molecular biomarkers (ERBB2, AKT, PTEN, HRR gene, CD274 amplification RB1, NF1, mutational process) in metastatic breast cancer, on the bases of the largest molecular profiling studies. These biomarkers will be classed according the level of evidence and, if possible, the ESCAT (ESMO) classification. Finally, we will provide some perspective on development in clinical practice for the main biomarkers.
Collapse
Affiliation(s)
- Benjamin Verret
- Medical Oncology Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
- INSERM Unit U981, Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Michele Bottosso
- INSERM Unit U981, Gustave Roussy Cancer Campus, 94805 Villejuif, France
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Sofia Hervais
- INSERM Unit U981, Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Barbara Pistilli
- Medical Oncology Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| |
Collapse
|
189
|
van der Wiel AMA, Schuitmaker L, Cong Y, Theys J, Van Hoeck A, Vens C, Lambin P, Yaromina A, Dubois LJ. Homologous Recombination Deficiency Scar: Mutations and Beyond-Implications for Precision Oncology. Cancers (Basel) 2022; 14:cancers14174157. [PMID: 36077694 PMCID: PMC9454578 DOI: 10.3390/cancers14174157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 02/05/2023] Open
Abstract
Homologous recombination deficiency (HRD) is a prevalent in approximately 17% of tumors and is associated with enhanced sensitivity to anticancer therapies inducing double-strand DNA breaks. Accurate detection of HRD would therefore allow improved patient selection and outcome of conventional and targeted anticancer therapies. However, current clinical assessment of HRD mainly relies on determining germline BRCA1/2 mutational status and is insufficient for adequate patient stratification as mechanisms of HRD occurrence extend beyond functional BRCA1/2 loss. HRD, regardless of BRCA1/2 status, is associated with specific forms of genomic and mutational signatures termed HRD scar. Detection of this HRD scar might therefore be a more reliable biomarker for HRD. This review discusses and compares different methods of assessing HRD and HRD scar, their advances into the clinic, and their potential implications for precision oncology.
Collapse
Affiliation(s)
- Alexander M. A. van der Wiel
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Lesley Schuitmaker
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ying Cong
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jan Theys
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Arne Van Hoeck
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Conchita Vens
- Institute of Cancer Science, University of Glasgow, Glasgow G61 1BD, Scotland, UK
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Correspondence:
| |
Collapse
|
190
|
Abstract
Triple negative breast cancer (TNBC) is characterized by the lack of estrogen and progesterone receptor expression and lacks HER2 overexpression or gene amplification. It accounts for 10–15% of incident breast cancers and carries the worst prognosis. TNBC is overrepresented among Black and pre-menopausal women and is associated with significant psychological and treatment-related burdens, including financial toxicity. Like other breast cancers, TNBC is biologically heterogeneous, leading to diverse clinical and epidemiological behaviors, however, unlike the other clinical subtypes, in TNBC we still lack tumor-specific targeted therapy. Early TNBC outcomes have improved due to the intensification of therapies, including improvements in polychemotherapy and the addition of immunotherapy. Future efforts are needed to identify targetable aberrations for specific drug therapy, prevent immune evasion, and increase social-economic support. Given that the name TNBC illustrates its lack of specifically targeted and effective therapy, we look forward to being able to retire the name in favor of a group of targetable entities within what is now called “TNBC”.
Collapse
|
191
|
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: 8] [Impact Index Per Article: 2.7] [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
|
192
|
Bai R, Jia L, Gao Y, Sun X, Chen N, Lv Z, Cui J. Targeted therapy combined with immunotherapy in patients with breast infiltrating ductal carcinoma with axillary lymph node metastasis of metaplastic SCC. Thorac Cancer 2022; 13:2799-2807. [PMID: 35975338 PMCID: PMC9527170 DOI: 10.1111/1759-7714.14602] [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: 06/10/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/02/2022] Open
Abstract
At present, the clinicopathological features, optimal treatment patterns, and prognosis of breast metaplastic squamous cell carcinoma (SCC) are not fully understood and are still controversial. Here, we report a 56‐year‐old female patient with breast infiltrating ductal carcinoma with axillary lymph node metastasis of metaplastic SCC admitted to our hospital. Their homology was clarified by comparing the gene mutation results of the two lesions, that is, the axillary lymph node lesion was a metastasis of breast metaplastic SCC. We treated the patient with Poly ADP‐ribose Polymerase (PARP) inhibitors in combination with immune checkpoint inhibitors (ICIs) and found that she could achieve clinical benefit from the combination regimen. We reported a successful diagnosis and treatment of this rare refractory disease and reviewed the literature on the characteristics, pathogenesis, and advances in the diagnosis and treatment of breast metaplastic SCC.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lin Jia
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yangyang Gao
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Sun
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Naifei Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zheng Lv
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
193
|
Functional assessment of missense variants of uncertain significance in the cancer susceptibility gene PALB2. NPJ Breast Cancer 2022; 8:86. [PMID: 35853885 PMCID: PMC9296472 DOI: 10.1038/s41523-022-00454-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Germline PALB2 pathogenic variants are associated with an increased lifetime risk for breast, pancreatic, and ovarian cancer. However, the interpretation of the pathogenicity of numerous PALB2 missense variants of uncertain significance (VUSs) identified in germline genetic testing remains a challenge. Here we selected ten potentially pathogenic PALB2 VUSs identified in 2279 Chinese patients with breast cancer and evaluated their impacts on PALB2 function by systematic functional assays. We showed that three PALB2 VUSs p.K16M [c.47 A > T], p.L24F [c.72 G > C], and p.L35F [c.103 C > T] in the coiled-coil domain impaired PALB2-mediated homologous recombination. The p.L24F and p.L35F variants partially disrupted BRCA1-PALB2 interactions, reduced RAD51 foci formation in response to DNA damage, abrogated ionizing radiation-induced G2/M checkpoint maintenance, and conferred increased sensitivity to olaparib and cisplatin. The p.K16M variant presented mild effects on BRCA1-PALB2 interactions and RAD51 foci formation. Altogether, we identify two novel PALB2 VUSs, p.L24F and p.L35F, that compromise PALB2 function and may increase cancer risk. These two variants display marked olaparib and cisplatin sensitivity and may help predict response to targeted therapy in the clinical treatment of patients with these variants.
Collapse
|
194
|
Functions of Breast Cancer Predisposition Genes: Implications for Clinical Management. Int J Mol Sci 2022; 23:ijms23137481. [PMID: 35806485 PMCID: PMC9267387 DOI: 10.3390/ijms23137481] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Approximately 5–10% of all breast cancer (BC) cases are caused by germline pathogenic variants (GPVs) in various cancer predisposition genes (CPGs). The most common contributors to hereditary BC are BRCA1 and BRCA2, which are associated with hereditary breast and ovarian cancer (HBOC). ATM, BARD1, CHEK2, PALB2, RAD51C, and RAD51D have also been recognized as CPGs with a high to moderate risk of BC. Primary and secondary cancer prevention strategies have been established for HBOC patients; however, optimal preventive strategies for most hereditary BCs have not yet been established. Most BC-associated CPGs participate in DNA damage repair pathways and cell cycle checkpoint mechanisms, and function jointly in such cascades; therefore, a fundamental understanding of the disease drivers in such cascades can facilitate the accurate estimation of the genetic risk of developing BC and the selection of appropriate preventive and therapeutic strategies to manage hereditary BCs. Herein, we review the functions of key BC-associated CPGs and strategies for the clinical management in individuals harboring the GPVs of such genes.
Collapse
|
195
|
Neoadjuvant study of niraparib in patients with HER2-negative, BRCA-mutated, resectable breast cancer. NATURE CANCER 2022; 3:927-931. [PMID: 35788722 PMCID: PMC9402431 DOI: 10.1038/s43018-022-00400-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 05/20/2022] [Indexed: 12/20/2022]
Abstract
AbstractThis single-arm pilot study (NCT03329937) evaluated neoadjuvant niraparib antitumor activity and safety in patients with localized HER2-negative, BRCA-mutated breast cancer. Twenty-one patients received niraparib 200 mg once daily in 28-day cycles. After 2 cycles, tumor response (≥30% reduction from baseline) by MRI was 90.5% and 40.0% (6 of 15) of patients who received only niraparib (2–6 cycles) had pathological complete response; no new safety signals were identified. High niraparib intratumoral concentration was observed.
Collapse
|
196
|
Tomiczek-Szwiec J, Szwiec M, Falco M, Cybulski C, Wokolorczyk D, Jakubowska A, Gronwald J, Stawicka M, Godlewski D, Kilar E, Marczyk E, Siołek M, Wiśniowski R, Haus O, Sibilski R, Bodnar L, Sun P, Narod SA, Lubinski J, Huzarski T. The impact of oophorectomy on survival from breast cancer in patients with CHEK2 mutations. Br J Cancer 2022; 127:84-91. [PMID: 35256754 PMCID: PMC9276789 DOI: 10.1038/s41416-022-01770-1] [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: 09/22/2021] [Revised: 02/07/2022] [Accepted: 02/17/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND To estimate the impact of oophorectomy and other treatments on the survival of breast cancer patients with a CHEK2 mutation. METHODS Women with Stage I-III breast cancer who were treated at 17 hospitals in Poland were tested for four founder mutations in the CHEK2 gene. 974 women (10%) were positive for a CHEK2 mutation. Control patients without a CHEK2 mutation were selected from a database of patients treated over the same time period. Information on treatments received and distant recurrences were retrieved from medical records. Treatments included chemotherapy, hormonal therapy (tamoxifen) and radiation therapy. Oophorectomies were performed for the treatment of breast cancer or for benign conditions. Dates of death were obtained from the Polish Vital Statistics Registry. Causes of death were determined by medical record review. Predictors of survival were determined using the Cox proportional hazards model. RESULTS In all, 839 patients with a CHEK2 mutation were matched to 839 patients without a mutation. The mean follow-up was 12.0 years. The 15-year survival for CHEK2 carriers was 76.6% and the 15-year survival for non-carrier control patients was 78.8% (adjusted HR = 1.06; 95% CI: 0.84-1.34; P = 0.61). Among CHEK2 carriers, the 15-year survival for women who had an oophorectomy was 86.3% and for women who did not have an oophorectomy was 72.1% (adjusted HR = 0.59; 95% CI: 0.38-0.90; P = 0.02). Among controls, the 15-year survival for patients who had an oophorectomy was 84.5% and for women who did not have an oophorectomy was 77.6% (adjusted HR = 1.03; 95% CI: 0.66-1.61; P = 0.90). CONCLUSION Among women with breast cancer and a CHEK2 mutation, oophorectomy is associated with a reduced risk of death from breast cancer.
Collapse
Affiliation(s)
- Joanna Tomiczek-Szwiec
- Department of Histology, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Opole, Poland
| | - Marek Szwiec
- Department of Surgery and Oncology, University of Zielona Góra, Zyty 28 St, 65-046, Zielona Góra, Poland.,Department of Clinical Oncology, University Hospital in Zielona Góra, Zyty 26 St, 65-046, Zielona Góra, Poland
| | - Michal Falco
- Regional Oncology Hospital, Strzalowska 22, 71-730, Szczecin, Poland
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1 St, 71-252, Szczecin, Poland
| | - Dominika Wokolorczyk
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1 St, 71-252, Szczecin, Poland
| | - Anna Jakubowska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1 St, 71-252, Szczecin, Poland
| | - Jacek Gronwald
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1 St, 71-252, Szczecin, Poland
| | - Malgorzata Stawicka
- Department of Clinical Genetics and Pathology, University of Zielona Góra, Zyty 28 St, 65-046, Zielona Góra, Poland
| | | | - Ewa Kilar
- Department of Oncology, District Specialist Hospital, Leśna 27-29 St, 58-100, Świdnica, Poland
| | - Elzbieta Marczyk
- Department of Oncological Surgery, Regional Oncology Center, Gancarska 11 St, 31-115, Kraków, Poland
| | - Monika Siołek
- Holycross Cancer Center, Artwińskiego 3 St, 25-734, Kielce, Poland
| | - Rafal Wiśniowski
- Regional Oncology Hospital, Wyzwolenia 18 St, 43-300, Bielsko Biała, Poland
| | - Olga Haus
- Department of Clinical Genetics, Collegium Medicum, Nicolaus Copernicus University, Jagiellońska 13 St, 85-067, Bydgoszcz, Poland
| | - Robert Sibilski
- Department of Surgery and Oncology, University of Zielona Góra, Zyty 28 St, 65-046, Zielona Góra, Poland.,Oncology Diagnostic Center, Wazów 42 St, 65-044, Zielona Góra, Poland
| | - Lubomir Bodnar
- Department of Oncology and Immuno-oncology, School of Medicine, Collegium Medicum, Uniwersity of Warmia and Mazury in Olsztyn, Warszawska 30 St, 10-082, Olsztyn, Poland
| | - Ping Sun
- Women's College Research Institute, Toronto, Ontario, M5S 1B2, Canada
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario, M5S 1B2, Canada.
| | - Jan Lubinski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1 St, 71-252, Szczecin, Poland
| | - Tomasz Huzarski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Unii Lubelskiej 1 St, 71-252, Szczecin, Poland.,Department of Clinical Genetics and Pathology, University of Zielona Góra, Zyty 28 St, 65-046, Zielona Góra, Poland
| | | |
Collapse
|
197
|
Kalinowski L, Viale G, Domchek S, Tutt A, Lucas PC, Lakhani SR. The increasing importance of pathology in modern clinical trial conduct: OlympiA as a case in point. Pathology 2022; 54:511-516. [PMID: 35778289 DOI: 10.1016/j.pathol.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022]
Affiliation(s)
- Lauren Kalinowski
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Qld, Australia; Department of Histopathology, Sullivan Nicolaides Pathology, Bowen Hills, Brisbane, Qld, Australia
| | - Giuseppe Viale
- European Institute of Oncology IRCCS, University of Milan, Milan, Italy
| | - Susan Domchek
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Tutt
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK; Breast Cancer Now Unit, The School of Cancer Studies and Pharmaceutical Science, King's College London, London, UK
| | - Peter C Lucas
- University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sunil R Lakhani
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Qld, Australia; Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, Brisbane, Qld, Australia.
| |
Collapse
|
198
|
Henry NL, Somerfield MR, Dayao Z, Elias A, Kalinsky K, McShane LM, Moy B, Park BH, Shanahan KM, Sharma P, Shatsky R, Stringer-Reasor E, Telli M, Turner NC, DeMichele A. Biomarkers for Systemic Therapy in Metastatic Breast Cancer: ASCO Guideline Update. J Clin Oncol 2022; 40:3205-3221. [PMID: 35759724 DOI: 10.1200/jco.22.01063] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE To update the ASCO biomarkers to guide systemic therapy for metastatic breast cancer (MBC) guideline. METHODS An Expert Panel conducted a systematic review to identify randomized clinical trials and prospective-retrospective studies from January 2015 to January 2022. RESULTS The search identified 19 studies informing the evidence base. RECOMMENDATIONS Candidates for a regimen with a phosphatidylinositol 3-kinase inhibitor and hormonal therapy should undergo testing for PIK3CA mutations using next-generation sequencing of tumor tissue or circulating tumor DNA (ctDNA) in plasma to determine eligibility for alpelisib plus fulvestrant. If no mutation is found in ctDNA, testing in tumor tissue, if available, should be used. Patients who are candidates for poly (ADP-ribose) polymerase (PARP) inhibitor therapy should undergo testing for germline BRCA1 and BRCA2 pathogenic or likely pathogenic mutations to determine eligibility for a PARP inhibitor. There is insufficient evidence for or against testing for a germline PALB2 pathogenic variant to determine eligibility for PARP inhibitor therapy in the metastatic setting. Candidates for immune checkpoint inhibitor therapy should undergo testing for expression of programmed cell death ligand-1 in the tumor and immune cells to determine eligibility for treatment with pembrolizumab plus chemotherapy. Candidates for an immune checkpoint inhibitor should also undergo testing for deficient mismatch repair/microsatellite instability-high to determine eligibility for dostarlimab-gxly or pembrolizumab, as well as testing for tumor mutational burden. Clinicians may test for NTRK fusions to determine eligibility for TRK inhibitors. There are insufficient data to recommend routine testing of tumors for ESR1 mutations, for homologous recombination deficiency, or for TROP2 expression to guide MBC therapy selection. There are insufficient data to recommend routine use of ctDNA or circulating tumor cells to monitor response to therapy among patients with MBC.Additional information can be found at www.asco.org/breast-cancer-guidelines.
Collapse
Affiliation(s)
| | | | | | | | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA
| | | | | | - Ben Ho Park
- Vanderbilt-Ingram Cancer Center, Nashville, TN
| | | | | | - Rebecca Shatsky
- University of California, San Diego School of Medicine, La Jolla, CA
| | | | | | | | | |
Collapse
|
199
|
Dixon-Douglas J, Loibl S, Denkert C, Telli M, Loi S. Integrating Immunotherapy Into the Treatment Landscape for Patients With Triple-Negative Breast Cancer. Am Soc Clin Oncol Educ Book 2022; 42:1-13. [PMID: 35649211 DOI: 10.1200/edbk_351186] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive histologic subtype of breast cancer for which, until recently, treatment options have been limited to chemotherapy. In recent years, an improved understanding of the importance of tumor-infiltrating lymphocytes and the tumor microenvironment in TNBC has led to investigation of immune checkpoint inhibitors for treatment. There is now evidence from several randomized controlled trials that supports the addition of immune checkpoint inhibitors to first-line treatment of advanced TNBC and to neoadjuvant chemotherapy for stage II-III TNBC. In parallel, the PARP inhibitors have emerged as a targeted therapy option for patients with HER2-negative breast cancer harboring mutations in BRCA1, BRCA2, and PALB2. Here, we review the recent clinical trials that inform the integration of immune checkpoint inhibitors into treatments for TNBC and discuss ongoing challenges-including patient selection, management of resistance to post-checkpoint inhibitor therapy, and combining immunotherapy with targeted therapies, including PARP inhibitors.
Collapse
Affiliation(s)
- Julia Dixon-Douglas
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Sibylle Loibl
- Goethe University Frankfurt, Germany.,Centre for Haematology and Oncology, Bethanein, Frankfurt, Germany.,German Breast Group, Neu-Isenburg, Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps-Universität Marburg and University Hospital of Giessen and Marburg, Marburg, Germany
| | - Melinda Telli
- Division of Medical Oncology, Stanford University School of Medicine, Stanford, CA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
200
|
Welslau M, Müller V, Lüftner D, Schütz F, Stickeler E, Fasching PA, Janni W, Thomssen C, Witzel I, Fehm TN, Belleville E, Bader S, Seitz K, Untch M, Thill M, Tesch H, Ditsch N, Lux MP, Aktas B, Banys-Paluchowski M, Schneeweiss A, Harbeck N, Würstlein R, Hartkopf AD, Wöckel A, Seliger B, Massa C, Kolberg HC. Update Breast Cancer 2022 Part 1 - Early Stage Breast Cancer. Geburtshilfe Frauenheilkd 2022; 82:580-589. [PMID: 35903719 PMCID: PMC9315400 DOI: 10.1055/a-1811-6106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 10/26/2022] Open
Abstract
Evidence relating to the treatment of breast cancer patients with early-stage disease has increased significantly in the past year. Abemaciclib, olaparib, and pembrolizumab are new drugs with good efficacy in the relevant patient groups. However, some questions remain unanswered. In particular, it remains unclear which premenopausal patients with hormone receptor-positive breast cancer should be spared unnecessary treatment. The question of the degree to which chemotherapy exerts a direct cytotoxic effect on the tumor or reduces ovarian function through chemotherapy could be of key importance. This group of patients could potentially be spared chemotherapy. New, previously experimental biomarker analysis methods, such as spatial analysis of gene expression (spatial transcriptomics) are gradually finding their way into large randomized phase III trials, such as the NeoTRIP trial. This in turn leads to a better understanding of the predictive factors of new therapies, for example immunotherapy. This review summarizes the scientific innovations from recent congresses such as the San Antonio Breast Cancer Symposium 2021 but also from recent publications.
Collapse
Affiliation(s)
| | - Volkmar Müller
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Diana Lüftner
- Charité University Hospital, Department of Hematology, Oncology and Tumour Immunology, University Medicine Berlin, Berlin, Germany
| | - Florian Schütz
- Gynäkologie und Geburtshilfe, Diakonissen-Stiftungs-Krankenhaus Speyer, Speyer, Germany
| | - Elmar Stickeler
- Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Aachen, Germany
| | - Peter A. Fasching
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Christoph Thomssen
- Department of Gynaecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Isabell Witzel
- Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
| | - Tanja N. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Simon Bader
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Katharina Seitz
- Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen,
Germany
| | - Michael Untch
- Clinic for Gynecology and Obstetrics, Breast Cancer Center, Genecologic Oncology Center, Helios Klinikum Berlin Buch, Berlin, Germany
| | - Marc Thill
- Agaplesion Markus Krankenhaus, Department of Gynecology and Gynecological Oncology, Frankfurt am Main
| | - Hans Tesch
- Oncology Practice at Bethanien Hospital, Frankfurt am Main, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, Germany
| | - Michael P. Lux
- Klinik für Gynäkologie und Geburtshilfe, Frauenklinik St. Louise, Paderborn, St. Josefs-Krankenhaus, Salzkotten, St. Vincenz Krankenhaus GmbH, Germany
| | - Bahriye Aktas
- Klinik und Poliklinik für Gynäkologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Maggie Banys-Paluchowski
- Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Nadia Harbeck
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
| | - Rachel Würstlein
- Breast Center, Department of Gynecology and Obstetrics and CCC Munich LMU, LMU University Hospital, Munich, Germany
| | - Andreas D. Hartkopf
- Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Barbara Seliger
- Martin-Luther-Universitat Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany
| | - Chiara Massa
- Martin-Luther-Universitat Halle-Wittenberg, Institute of Medical Immunology, Halle (Saale), Germany
| | | |
Collapse
|