151
|
Singh V, Kumar K, Purohit D, Verma R, Pandey P, Bhatia S, Malik V, Mittal V, Rahman MH, Albadrani GM, Arafah MW, El-Demerdash FM, Akhtar MF, Saleem A, Kamel M, Najda A, Abdel-Daim MM, Kaushik D. Exploration of therapeutic applicability and different signaling mechanism of various phytopharmacological agents for treatment of breast cancer. Biomed Pharmacother 2021; 139:111584. [PMID: 34243623 DOI: 10.1016/j.biopha.2021.111584] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cancer is one of the most dreaded diseases characterized by uncontrolled proliferation of abnormal cells that occurs due to impairment of cell division and apoptosis process. Cancer is categorized into several types on the basis of affected organs and breast cancer (BC) is the most predominant cause of mortality among women. Although, several synthetic and semi-synthetic therapies have been developed for the treatment of BC but they exhibit numerous serious adverse effects therefore; pharmacological agents with fewer/no side effects need to be explored. Plants and phytoconstituents perhaps fulfill the aforementioned requirement and could serve as a potential and alternative therapy for BC treatment. The ongoing biomedical research, clinical trials and number of patents granted have further boosted the acceptance of the plants and plant-derived constituents in the effective treatment of BC. PURPOSE OF STUDY Various treatment strategies such as checkpoint inhibitors, targeting micro RNA, apoptotic pathway, BRCA-1 gene, P53 protein, P13K/Akt/mTOR pathway, notch signaling pathway, hedgehog/gli-1 signaling pathway, poly-ADP ribose polymerase inhibitors, mitogen-activated protein kinase inhibitors etc. are available for BC. In addition to these synthetic and semi-synthetic drug therapies, several natural constituents such as alkaloids, sesquiterpenes, polyphenols, flavonoids and diterpenoids from medicinal plants, vegetables and fruits are reported to possess promising anti-cancer activity. The purpose of the present review is to highlight the various signaling pathways through which plants/herbs show the anti-cancer potential especially against the BC. STUDY DESIGN The literature for the present study was collected from various databases such as Pubmed, Scopus, Chemical Abstracts, Medicinal and aromatic plant abstracts, Web of Science etc. The different patent databases were also reviewed for the anti-cancer (BC) potential of the particular herbs/plants and their formulations. RESULT AND CONCLUSION In this review, we have discussed the number of plants along with their patents of different herbal formulations which are being used for the treatment of BC and other types of cancers. We have also delineated the different signaling mechanisms through which they inhibit the growth of BC cells. In nutshell, we can conclude that large numbers of herbs or their extracts are reported for the treatment of BC. But still, there is further need for research in-depth to translate the use of natural products clinically BC treatment.
Collapse
Affiliation(s)
- Vandana Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India; University Institute of Pharmaceutical Sciences (UIPS), Chandigarh University, Mohali, Punjab, India
| | - Deepika Purohit
- Department of Pharmaceutical Sciences, Indira Gandhi University, Rewari 123401, Haryana, India
| | - Ravinder Verma
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Parijat Pandey
- Department of Pharmaceutical Sciences, Gurugram University, Gurugram
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University Haryana, Manesar, Panchgaon, Haryana 122412, India; Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Vinay Malik
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohammed W Arafah
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Fatma M El-Demerdash
- Department of Environmental Studies, Institute of Graduate Studies and Research, University of Alexandria, Alexandria, Egypt
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegtable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland.
| | - Mohamed M Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India.
| |
Collapse
|
152
|
Nguyen AAT, Arasu VA, Strand F, Li W, Onishi N, Gibbs J, Jones EF, Joe BN, Esserman LJ, Newitt DC, Hylton NM. Comparison of Segmentation Methods in Assessing Background Parenchymal Enhancement as a Biomarker for Response to Neoadjuvant Therapy. ACTA ACUST UNITED AC 2021; 6:101-110. [PMID: 32548286 PMCID: PMC7289261 DOI: 10.18383/j.tom.2020.00009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Breast parenchymal enhancement (BPE) has shown association with breast cancer risk and response to neoadjuvant treatment. However, BPE quantification is challenging, and there is no standardized segmentation method for measurement. We investigated the use of a fully automated breast fibroglandular tissue segmentation method to calculate BPE from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for use as a predictor of pathologic complete response (pCR) following neoadjuvant treatment in the I-SPY 2 TRIAL. In this trial, patients had DCE-MRI at baseline (T0), after 3 weeks of treatment (T1), after 12 weeks of treatment and between drug regimens (T2), and after completion of treatment (T3). A retrospective analysis of 2 cohorts was performed: one with 735 patients and another with a final cohort of 340 patients, meeting a high-quality benchmark for segmentation. We evaluated 3 subvolumes of interest segmented from bilateral T1-weighted axial breast DCE-MRI: full stack (all axial slices), half stack (center 50% of slices), and center 5 slices. The differences between methods were assessed, and a univariate logistic regression model was implemented to determine the predictive performance of each segmentation method. The results showed that the half stack method provided the best compromise between sampling error from too little tissue and inclusion of incorrectly segmented tissues from extreme superior and inferior regions. Our results indicate that BPE calculated using the half stack segmentation approach has potential as an early biomarker for response to treatment in the hormone receptor–negative and human epidermal growth factor receptor 2–positive subtype.
Collapse
Affiliation(s)
- Alex Anh-Tu Nguyen
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Vignesh A Arasu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA.,Department of Radiology, Kaiser Permanente Medical Center, Vallejo, CA
| | - Fredrik Strand
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden; and
| | - Wen Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Natsuko Onishi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Jessica Gibbs
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Ella F Jones
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Bonnie N Joe
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Laura J Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA
| | - David C Newitt
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Nola M Hylton
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
153
|
Li W, Newitt DC, Yun BL, Jones EF, Arasu V, Wilmes LJ, Gibbs J, Nguyen AAT, Onishi N, Kornak J, Joe BN, Esserman LJ, Hylton NM. Tumor Sphericity Predicts Response in Neoadjuvant Chemotherapy for Invasive Breast Cancer. ACTA ACUST UNITED AC 2021; 6:216-222. [PMID: 32548299 PMCID: PMC7289243 DOI: 10.18383/j.tom.2020.00016] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This retrospective study examined magnetic resonance imaging (MRI)–derived tumor sphericity (SPH) as a quantitative measure of breast tumor morphology, and investigated the association between SPH and reader-assessed morphological pattern (MP). In addition, association of SPH with pathologic complete response was evaluated in patients enrolled in an adaptively randomized clinical trial designed to rapidly identify new agents for breast cancer. All patients underwent MRI examinations at multiple time points during the treatment. SPH values from pretreatment (T0) and early-treatment (T1) were investigated in this study. MP on T0 dynamic contrast-enhanced MRI was ranked from 1 to 5 in 220 patients. Mean SPH values decreased with the increased order of MP. SPH was higher in patients with pathologic complete response than in patients without (difference at T0: 0.04, 95% confidence interval [CI]: 0.02–0.05, P < .001; difference at T1: 0.03, 95% CI: 0.02–0.04, P < .001). The area under the receiver operating characteristic curve was estimated as 0.61 (95% CI, 0.57–0.65) at T0 and 0.58 (95% CI, 0.55–0.62) at T1. When the analysis was performed by cancer subtype defined by hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) status, highest area under the receiver operating characteristic curve were observed in HR−/HER2+: 0.67 (95% CI, 0.54–0.80) at T0, and 0.63 (95% CI, 0.51–0.76) at T1. Tumor SPH showed promise to quantify MRI MPs and as a biomarker for predicting treatment outcome at pre- or early-treatment time points.
Collapse
Affiliation(s)
- Wen Li
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - David C Newitt
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Bo La Yun
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA.,Department of Radiology, Seoul National University Bundang Hospital, Seoul
| | - Ella F Jones
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Vignesh Arasu
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Lisa J Wilmes
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Jessica Gibbs
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Alex Anh-Tu Nguyen
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Natsuko Onishi
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - John Kornak
- Departments of Epidemiology and Biostatistics; and
| | - Bonnie N Joe
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Laura J Esserman
- Surgery, University of California, San Francisco, San Francisco, CA
| | - Nola M Hylton
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
154
|
Potter DA, Thomas A, Rugo HS. A Neoadjuvant Chemotherapy Trial for Early Breast Cancer is Impacted by COVID-19: Addressing Vaccination and Cancer Trials Through Education, Equity, and Outcomes. Clin Cancer Res 2021; 27:4486-4490. [PMID: 34108186 DOI: 10.1158/1078-0432.ccr-21-1133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/14/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022]
Abstract
While COVID-19 vaccine distribution has addressed vulnerabilities related to age and comorbidities, there is a need to ensure vaccination of patients with cancer receiving experimental and routine treatment, where interruption of treatment by infection is likely to result in inferior outcomes. Among patients with cancer, those undergoing neoadjuvant chemotherapy (NAC) or adjuvant chemotherapy (Adj chemo) for early breast cancer (EBC) are at particularly high risk for inferior outcomes, in part, because optimal timing of chemotherapy is essential for promoting distant disease-free survival. COVID-19 data from the ongoing multicenter I-SPY 2 trial of NAC for EBC provides a window into the magnitude of the problem of treatment interruption, not only for the trial itself but also for routine Adj chemo. In the I-SPY 2 trial, 4.5% of patients had disruption of therapy by COVID-19, prior to wide vaccine availability, suggesting that nationally up to 5,700 patients with EBC were at risk for adverse outcomes from COVID-19 infection in 2020. To address this problem, vaccine education and public engagement are essential to overcome hesitancy, while equity of distribution is needed to address access. To accomplish these goals, healthcare organizations (HCO) need to not only call out disinformation but also engage the public with vaccine education and find common ground for vaccine acceptance, while partnering with state/local governments to improve efficiency of vaccine distribution. These approaches are important to improve trial access and to reduce susceptibility to COVID-19, as the pandemic could continue to impact access to clinical trials and routine cancer treatment.
Collapse
Affiliation(s)
- David A Potter
- University of Minnesota Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, Minneapolis, Minnesota.
| | - Alexandra Thomas
- Wake Forest Baptist Health Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, California
| |
Collapse
|
155
|
Oh SY, Rahman S, Sparano JA. Perspectives on PARP inhibitors as pharmacotherapeutic strategies for breast cancer. Expert Opin Pharmacother 2021; 22:981-1003. [PMID: 33646064 PMCID: PMC9047307 DOI: 10.1080/14656566.2021.1876662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/12/2021] [Indexed: 12/17/2022]
Abstract
Introduction Approximately 10% of all breast cancer cases occur in individuals who have germline pathogenic variants of the BRCA 1, BRCA 2, and other genes associated with impaired DNA damage repair that is associated with an increased risk of breast, ovarian, and other cancers. Inhibitors of poly-ADP ribose polymerase (PARP) induce synthetic lethality in cancer cells harboring such pathogenic variants.Area covered In this review, the authors review the mechanisms of action, antitumor activity, and adverse events associated with PARP inhibitors for the treatment of advanced breast cancer. The authors then summarize the area and provide their expert perspectives on the area.Expert opinion Two PARP inhibitors are approved in metastatic breast cancer, including olaparib and talozaparib. Both agents were approved based on phase III trials demonstrating that they were associated with improved progression-free survival compared with treatment of physician's choice in patients receiving second-third line therapy for locally advanced, inoperable, or metastatic breast cancer in patients with germline pathogenic BRCA 1 or BRCA2 variants.
Collapse
Affiliation(s)
- Sun Young Oh
- Department of Hematology and Oncology, Montefiore-Einstein center for cancer care, Albert Einstein College of Medicine, Bronx, NY
| | - Shafia Rahman
- Department of Hematology and Oncology, Montefiore-Einstein center for cancer care, Albert Einstein College of Medicine, Bronx, NY
| | - Joseph A Sparano
- Department of Hematology and Oncology, Montefiore-Einstein center for cancer care, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
156
|
Breaking the therapeutic ceiling in drug development in ulcerative colitis. Lancet Gastroenterol Hepatol 2021; 6:589-595. [PMID: 34019798 DOI: 10.1016/s2468-1253(21)00065-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Increased knowledge of the intricate pathogenesis of ulcerative colitis has triggered an advance in drug development during the past two decades, resulting in the advent of several biological agents and small-molecule therapies. Although the increase in therapeutic options is positive, remission rates of patients with ulcerative colitis given new therapeutic agents in induction trials remain at a modest 20-30%, seemingly facing a so-called therapeutic ceiling. This therapeutic ceiling requires a critical appraisal and highlights the need for alternative strategies for drug development. In this Review, we objectively itemise the boundaries of therapeutic efficacy in ulcerative colitis, provide possible explanations for the shortcomings of current strategies, and propose solutions to achieve better therapeutic outcomes in ulcerative colitis.
Collapse
|
157
|
Tene K, Kalyan Kumar M, Basveshwar G, Eswara Rao P, Jagadeesh Kumar G, kumar P, Pemmaraju DB, Murty USN, Gogoi R, Naidu VGM. Polyphenolic-Rich Compounds From Dillenia pentagyna (Roxb.) Attenuates the Doxorubicin-Induced Cardiotoxicity: A High-Frequency Ultrasonography Assisted Approach. Front Pharmacol 2021; 12:624706. [PMID: 34079455 PMCID: PMC8166202 DOI: 10.3389/fphar.2021.624706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/15/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular complications are the foremost concern in patients undergoing anticancer therapy. There is an unmet need to address the problems arising from the drug-induced toxicity for the long-term benefit of the patients undergoing chemotherapy. Alternative medicines are gaining their prosperity in addressing the various drug-induced organ toxicity. Dillenia pentagyna Roxb (DP) is an ethnomedicinal plant rich in flavonoids and phenolic contents. In India & Nepal, DP is a common ingredient of traditional medicines used to treat multiple ailments like inflammation, cancer, and diabetes. However, its protective role against doxorubicin (Dox) induced cardiotoxicity remains unexplored. Herein, we investigated the potential effects of various extracts/fractions obtained from the DP's bark against Dox-induced cardiotoxicity, both in-vitro and in-vivo. The anti-oxidant content of the extracts/fractions was evaluated by using DPPH, ABTS and FRAP chemical assays. The results indicated that the hydroalcoholic (HA) extract of DP has intense anti-oxidant potential. Further fractionation of DP revealed that the phenolic-rich fraction (F1) has a high anti-oxidant potential. The protective effect of extract/fraction was also investigated in the H9c2 cell line following the Dox-induced cardiotoxicity model. We observed that the pre-treatment of extract/fraction in cardiomyocytes had exhibited increased cell viability. Fluorescence-based chemical assays indicated a decreased ROS levels in the treated groups in comparison to the Dox control group. The effect of DP was evaluated further in balb/c mice by the Dox-induced cardiotoxicity model. Non-invasive techniques like high-frequency ultrasonography and electrocardiogram revealed that the mice pre-treated with DP had improved cardiac functionality (left ventricular ejection fraction and stroke volume) and normalized the electrocardiograms compared to the Dox control group. Further, biochemical analysis with the cardiac tissues revealed that the cytoprotective proteins like HO-1, SOD-2, and Nrf-2 were elevated in the DP treated groups compared to the Dox control group. Overall, our results suggested that the bioactive extract/fractions of DP helped alleviate the Dox-induced cardiotoxicity. LC-QTOF-ESI-MS analysis of DP and F1 indicated that polyphenolic anti-oxidant compounds like gallic acid, syringic acid, and sinapic acid could be responsible for the potent -cardioprotective effect. Future understanding of the pharmacokinetics and pharmacodynamic parameters can help translate from the bench to the bedside.
Collapse
Affiliation(s)
- Kalyani Tene
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Guwahati, Assam, India
| | - M. Kalyan Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| | - G. Basveshwar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| | - P. Eswara Rao
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| | - G. Jagadeesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| | - Pramod kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research Guwahati, Assam, India
| | - Deepak B. Pemmaraju
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| | - U. S. N. Murty
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| | - Ranadeep Gogoi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Guwahati, Assam, India
- Biological Sciences and Technology Department, CSIR-North-East Institute of Science and Technology, Assam, India
| | - V. G. M. Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Guwahati, Assam, India
| |
Collapse
|
158
|
Miglietta F, Dieci MV, Griguolo G, Guarneri V. Neoadjuvant approach as a platform for treatment personalization: focus on HER2-positive and triple-negative breast cancer. Cancer Treat Rev 2021; 98:102222. [PMID: 34023642 DOI: 10.1016/j.ctrv.2021.102222] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 05/09/2021] [Indexed: 10/21/2022]
Abstract
The neoadjuvant setting provides unquestionable clinical benefits for high-risk breast cancer (BC) patients, mainly in terms of expansion of locoregional treatment options and prognostic stratification. Additionally, it is also emerging as a strategical tool in the research field. In the present review, by focusing on HER2-positive and triple-negative subtypes, we examined the role of the neoadjuvant setting as a research platform to facilitate and rationalize the placement of escalation strategies, promote the adoption of biomarker-driven approaches for the investigation of de-escalated treatments, and foster the conduction of comprehensive translational analyses, thus ultimately aiming at pursuing treatment personalization. The solid prognostic role of pathologic complete response after neoadjuvant therapy, and its use as a surrogate endpoint to accelerate the drug approval process were discussed. In this context, available data on escalated treatment strategies capable of enhancing pathologic complete response (pCR) rate or improving prognosis of patients with residual disease (RD) after neoadjuvant treatment, were comprehensively reviewed. We also summarized evidence regarding the possibility of obtaining pCR with de-escalated strategies, with particular emphasis on the role of biomarker-driven approaches for patient selection. Pitfalls of the dichotomy of pCR/RD were also deepened, and data on alternative/complementary biomarkers with a possible clinical relevance in this regard were reviewed.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.
| | - Gaia Griguolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| |
Collapse
|
159
|
Korde LA, Somerfield MR, Carey LA, Crews JR, Denduluri N, Hwang ES, Khan SA, Loibl S, Morris EA, Perez A, Regan MM, Spears PA, Sudheendra PK, Symmans WF, Yung RL, Harvey BE, Hershman DL. Neoadjuvant Chemotherapy, Endocrine Therapy, and Targeted Therapy for Breast Cancer: ASCO Guideline. J Clin Oncol 2021; 39:1485-1505. [PMID: 33507815 PMCID: PMC8274745 DOI: 10.1200/jco.20.03399] [Citation(s) in RCA: 579] [Impact Index Per Article: 144.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/04/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To develop guideline recommendations concerning optimal neoadjuvant therapy for breast cancer. METHODS ASCO convened an Expert Panel to conduct a systematic review of the literature on neoadjuvant therapy for breast cancer and provide recommended care options. RESULTS A total of 41 articles met eligibility criteria and form the evidentiary basis for the guideline recommendations. RECOMMENDATIONS Patients undergoing neoadjuvant therapy should be managed by a multidisciplinary care team. Appropriate candidates for neoadjuvant therapy include patients with inflammatory breast cancer and those in whom residual disease may prompt a change in therapy. Neoadjuvant therapy can also be used to reduce the extent of local therapy or reduce delays in initiating therapy. Although tumor histology, grade, stage, and estrogen, progesterone, and human epidermal growth factor receptor 2 (HER2) expression should routinely be used to guide clinical decisions, there is insufficient evidence to support the use of other markers or genomic profiles. Patients with triple-negative breast cancer (TNBC) who have clinically node-positive and/or at least T1c disease should be offered an anthracycline- and taxane-containing regimen; those with cT1a or cT1bN0 TNBC should not routinely be offered neoadjuvant therapy. Carboplatin may be offered to patients with TNBC to increase pathologic complete response. There is currently insufficient evidence to support adding immune checkpoint inhibitors to standard chemotherapy. In patients with hormone receptor (HR)-positive (HR-positive), HER2-negative tumors, neoadjuvant chemotherapy can be used when a treatment decision can be made without surgical information. Among postmenopausal patients with HR-positive, HER2-negative disease, hormone therapy can be used to downstage disease. Patients with node-positive or high-risk node-negative, HER2-positive disease should be offered neoadjuvant therapy in combination with anti-HER2-positive therapy. Patients with T1aN0 and T1bN0, HER2-positive disease should not be routinely offered neoadjuvant therapy.Additional information is available at www.asco.org/breast-cancer-guidelines.
Collapse
Affiliation(s)
- Larissa A Korde
- Clinical Investigations Branch, CTEP, DCTD, National Cancer Institute, Bethesda, MD
| | | | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | | | | | | | | | | | | - Alejandra Perez
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Plantation, FL
| | | | - Patricia A Spears
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | | | | | | | | - Dawn L Hershman
- Herbert Irving Comprehensive Cancer Center at Columbia University, New York, NY
| |
Collapse
|
160
|
Israel E, Denlinger LC, Bacharier LB, LaVange LM, Moore WC, Peters MC, Georas SN, Wright RJ, Mauger DT, Noel P, Akuthota P, Bach J, Bleecker ER, Cardet JC, Carr TF, Castro M, Cinelli A, Comhair SAA, Covar RA, Alexander LC, DiMango EA, Erzurum SC, Fahy JV, Fajt ML, Gaston BM, Hoffman EA, Holguin F, Jackson DJ, Jain S, Jarjour NN, Ji Y, Kenyon NJ, Kosorok MR, Kraft M, Krishnan JA, Kumar R, Liu AH, Liu MC, Ly NP, Marquis MA, Martinez FD, Moy JN, O'Neal WK, Ortega VE, Peden DB, Phipatanakul W, Ross K, Smith LJ, Szefler SJ, Teague WG, Tulchinsky AF, Vijayanand P, Wechsler ME, Wenzel SE, White SR, Zeki AA, Ivanova A. PrecISE: Precision Medicine in Severe Asthma: An adaptive platform trial with biomarker ascertainment. J Allergy Clin Immunol 2021; 147:1594-1601. [PMID: 33667479 PMCID: PMC8113113 DOI: 10.1016/j.jaci.2021.01.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Severe asthma accounts for almost half the cost associated with asthma. Severe asthma is driven by heterogeneous molecular mechanisms. Conventional clinical trial design often lacks the power and efficiency to target subgroups with specific pathobiological mechanisms. Furthermore, the validation and approval of new asthma therapies is a lengthy process. A large proportion of that time is taken by clinical trials to validate asthma interventions. The National Institutes of Health Precision Medicine in Severe and/or Exacerbation Prone Asthma (PrecISE) program was established with the goal of designing and executing a trial that uses adaptive design techniques to rapidly evaluate novel interventions in biomarker-defined subgroups of severe asthma, while seeking to refine these biomarker subgroups, and to identify early markers of response to therapy. The novel trial design is an adaptive platform trial conducted under a single master protocol that incorporates precision medicine components. Furthermore, it includes innovative applications of futility analysis, cross-over design with use of shared placebo groups, and early futility analysis to permit more rapid identification of effective interventions. The development and rationale behind the study design are described. The interventions chosen for the initial investigation and the criteria used to identify these interventions are enumerated. The biomarker-based adaptive design and analytic scheme are detailed as well as special considerations involved in the final trial design.
Collapse
Affiliation(s)
- Elliot Israel
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass.
| | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | | | - Lisa M LaVange
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Wendy C Moore
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - Michael C Peters
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY
| | | | - David T Mauger
- Pennsylvania State University School of Medicine, Hershey, Pa
| | - Patricia Noel
- Division of Lung Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Praveen Akuthota
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Julia Bach
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Eugene R Bleecker
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | | | - Tara F Carr
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, Kan
| | | | | | | | - Laura Crotty Alexander
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | | | | | - John V Fahy
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | - Merritt L Fajt
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Benjamin M Gaston
- Wells Center for Pediatric Research, Indiana University, Indianapolis, Ind
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | | | - Daniel J Jackson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Sonia Jain
- Pulmonary Division, Department of Medicine, University of California-San Diego, La Jolla, Calif
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Yuan Ji
- Department of Health Studies, University of Chicago, Chicago, Ill
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Michael R Kosorok
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Monica Kraft
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - Jerry A Krishnan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Ill
| | | | - Andrew H Liu
- University of Colorado School of Medicine, Aurora, Colo; Children's Hospital Colorado, Aurora, Colo
| | - Mark C Liu
- Pulmonary and Critical Care Medicine, Department of Medicine, the Johns Hopkins University, Baltimore, Md
| | - Ngoc P Ly
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | - M Alison Marquis
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Fernando D Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Ariz
| | - James N Moy
- Rush University Medical Center, Chicago, Ill
| | - Wanda K O'Neal
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC
| | - Victor E Ortega
- Wake Forest University School of Medicine, Winston-Salem, NC
| | - David B Peden
- Marsico Lung Institute, UNC CF Research Center, University of North Carolina, Chapel Hill, NC
| | | | - Kristie Ross
- UH Rainbow Babies and Children's Hospitals, Cleveland, Ohio
| | | | - Stanley J Szefler
- University of Colorado School of Medicine, Aurora, Colo; Children's Hospital Colorado, Aurora, Colo
| | - W Gerald Teague
- University of Virginia School of Medicine, Charlottesville, Va
| | | | | | - Michael E Wechsler
- National Jewish Health, Denver, Colo; University of Colorado School of Medicine, Aurora, Colo
| | - Sally E Wenzel
- University of Pittsburgh Asthma Institute, University of Pittsburgh, Pittsburgh, Pa
| | - Steven R White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Ill
| | - Amir A Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis School of Medicine, Davis, Calif
| | - Anastasia Ivanova
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
161
|
May Lee K, Jack Lee J. Evaluating Bayesian adaptive randomization procedures with adaptive clip methods for multi-arm trials. Stat Methods Med Res 2021; 30:1273-1287. [PMID: 33689524 PMCID: PMC7613973 DOI: 10.1177/0962280221995961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Bayesian adaptive randomization is a heuristic approach that aims to randomize more patients to the putatively superior arms based on the trend of the accrued data in a trial. Many statistical aspects of this approach have been explored and compared with other approaches; yet only a limited number of works has focused on improving its performance and providing guidance on its application to real trials. An undesirable property of this approach is that the procedure would randomize patients to an inferior arm in some circumstances, which has raised concerns in its application. Here, we propose an adaptive clip method to rectify the problem by incorporating a data-driven function to be used in conjunction with Bayesian adaptive randomization procedure. This function aims to minimize the chance of assigning patients to inferior arms during the early time of the trial. Moreover, we propose a utility approach to facilitate the selection of a randomization procedure. A cost that reflects the penalty of assigning patients to the inferior arm(s) in the trial is incorporated into our utility function along with all patients benefited from the trial, both within and beyond the trial. We illustrate the selection strategy for a wide range of scenarios.
Collapse
Affiliation(s)
- Kim May Lee
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - J Jack Lee
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
162
|
Taylor AM, Chan DLH, Tio M, Patil SM, Traina TA, Robson ME, Khasraw M. PARP (Poly ADP-Ribose Polymerase) inhibitors for locally advanced or metastatic breast cancer. Cochrane Database Syst Rev 2021; 4:CD011395. [PMID: 33886122 PMCID: PMC8092476 DOI: 10.1002/14651858.cd011395.pub2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Locally advanced and metastatic breast cancer remains a challenge to treat. With emerging study results, it is important to interpret the available clinical data and apply the evidence offering the most effective treatment to the right patient. Poly(ADP Ribose) Polymerase (PARP) inhibitors are a new class of drug and their role in the treatment of locally advanced and metastatic breast cancer is being established. OBJECTIVES To determine the efficacy, safety profile, and potential harms of Poly(ADP-Ribose) Polymerase (PARP) inhibitors in the treatment of patients with locally advanced or metastatic breast cancer. The primary outcome of interest was overall survival; secondary outcomes included progression-free survival, tumour response rate, quality of life, and adverse events. SEARCH METHODS On 8 June 2020, we searched the Cochrane Breast Cancer Group Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE via OvidSP, Embase via OvidSP, World Health Organization International Clinical Trials Registry Platform (WHO ICTRP) search portal and ClinicalTrials.gov. We also searched proceedings from the major oncology conferences as well as scanned reference lists from eligible publications and contacted corresponding authors of trials for further information, where needed. SELECTION CRITERIA We included randomised controlled trials on participants with locally advanced or metastatic breast cancer comparing 1) chemotherapy in combination with PARP inhibitors, compared to the same chemotherapy without PARP inhibitors or 2) treatment with PARP inhibitors, compared to treatment with other chemotherapy. We included studies that reported on our primary outcome of overall survival and secondary outcomes including progression-free survival, tumour response rate, quality of life, and adverse events. DATA COLLECTION AND ANALYSIS We used standard methodological procedures defined by Cochrane. Summary statistics for the endpoints used hazard ratios (HR) with 95% confidence intervals (CI) for overall survival and progression-free survival, and odds ratios (OR) for response rate (RR) and toxicity. MAIN RESULTS We identified 49 articles for qualitative synthesis, describing five randomised controlled trials that were included in the quantitative synthesis (meta-analysis). A sixth trial was assessed as eligible but had ended prematurely and no data were available for inclusion in our meta-analysis. Risk of bias was predominately low to unclear across all studies except in regards to performance bias (3/5 high risk) and detection bias for the outcomes of quality of life (2/2 high risk) and reporting of adverse events (3/5 high risk). High-certainty evidence shows there may be a small advantage in overall survival (HR 0.87, 95% CI 0.76 to 1.00; 4 studies; 1435 patients). High-certainty evidence shows that PARP inhibitors offer an improvement in PFS in locally advanced/metastatic HER2-negative, BRCA germline mutated breast cancer patients (HR 0.63, 95% CI 0.56 to 0.71; 5 studies; 1474 patients). There was no statistical heterogeneity for these outcomes. Subgroup analyses for PFS outcomes based on trial level data were performed for triple-negative breast cancer, hormone-positive and/or HER2-positive breast cancer, BRCA1 and BRCA2 germline mutations, and patients who had received prior chemotherapy for advanced breast cancer or not. The subgroup analyses showed a persistent PFS benefit regardless of the subgroup chosen. Pooled analysis shows PARP inhibitors likely result in a moderate improvement in tumour response rate compared to other treatment arms (66.9% vs 48.9%; RR 1.39, 95% CI 1.24 to 1.54; 5 studies; 1185 participants; moderate-certainty evidence). The most common adverse events reported across all five studies included neutropenia, anaemia and fatigue. Grade 3 or higher adverse events probably occur no less frequently in patients receiving PARP inhibitors (59.4% for PARP arm versus 64.5% for non-PARP arm, RR 0.98, 95% CI 0.91 to 1.04; 5 studies; 1443 participants; moderate-certainty evidence). Only two studies reported quality of life outcomes so this was not amenable to meta-analysis. However, both studies that did assess quality of life showed PARP inhibitors were superior compared to physician's choice of chemotherapy in terms of participant-reported outcomes. AUTHORS' CONCLUSIONS In people with locally advanced or metastatic HER2-negative, BRCA germline mutated breast cancer, PARP inhibitors offer an improvement in progression-free survival, and likely improve overall survival and tumour response rates. This systematic review provides evidence supporting the use of PARP inhibitors as part of the therapeutic strategy for breast cancer patients in this subgroup. The toxicity profile for PARP inhibitors is probably no worse than chemotherapy but more information is required regarding quality of life outcomes, highlighting the importance of collecting such data in future studies. Future studies should also be powered to detect clinically important differences in overall survival and could focus on the role of PARP inhibitors in other relevant breast cancer populations, including HER2-positive, BRCA-negative/homologous recombination repair-deficient and Programmed Death-Ligand 1 (PDL1) positive.
Collapse
Affiliation(s)
- Amelia M Taylor
- Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - David Lok Hang Chan
- Medical Oncology, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, Australia
| | - Martin Tio
- Medical Oncology, Royal North Shore Hospital, Sydney, Australia
| | - Sujata M Patil
- Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tiffany A Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Mustafa Khasraw
- NHMRC Clinical Trials Centre, The University of Sydney, Camperdown, Australia
| |
Collapse
|
163
|
McDonald ES, Pantel AR, Shah PD, Farwell MD, Clark AS, Doot RK, Pryma DA, Carlin SD. In vivo visualization of PARP inhibitor pharmacodynamics. JCI Insight 2021; 6:146592. [PMID: 33884961 PMCID: PMC8119179 DOI: 10.1172/jci.insight.146592] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/10/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND [18F]FluorThanatrace ([18F]FTT) is a radiolabeled poly (adenosine diphosphate-ribose) polymerase inhibitor (PARPi) that enables noninvasive quantification of PARP with potential to serve as a biomarker for patient selection for PARPi therapy. Here we report for the first time to our knowledge noninvasive in vivo visualization of drug-target engagement during PARPi treatment. METHODS Two single-arm, prospective, nonrandomized clinical trials were conducted at the University of Pennsylvania from May 2017 to March 2020. PARP expression in breast cancer was assessed in vivo via [18F]FTT PET before and after initiation of PARPi treatment and in vitro via [125I]KX1 (an analog of [18F]FTT) binding to surgically removed breast cancer. RESULTS Thirteen patients had baseline [18F]FTT PET. Nine of these then had resection and in vitro evaluation of [18F]FTT uptake with an analog and uptake was blocked with PARPi. Of the other 4 patients, 3 had [18F]FTT PET uptake, and all had uptake blocked with treatment with a therapeutic PARPi. Initial in vivo [18F]FTT tumor uptake ranged from undetectable to robust. Following initiation of PARPi therapy, [18F]FTT uptake was not detectable above background in all cases. In vitro tumor treatment with a PARPi resulted in 82% reduction in [125I]KX1 binding. CONCLUSION [18F]FTT noninvasively quantifies PARP-1 expression. Early results indicate ability to visualize PARPi drug-target engagement in vivo and suggest the utility of further study to test [18F]FTT PET as a predictive and pharmacodynamic biomarker. TRIAL REGISTRATION ClinicalTrials.gov identifiers NCT03083288 and NCT03846167. FUNDING Metavivor Translational Research Award, Susan G. Komen for the Cure (CCR 16376362), Department of Defense BC190315, and Abramson Cancer Center Breakthrough Bike Challenge. Human PARP inhibitor drug-target engagement can be visualized, with future possibilities including a precision diagnostic and drug discovery tool.
Collapse
Affiliation(s)
| | - Austin R Pantel
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, and
| | - Payal D Shah
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael D Farwell
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, and
| | - Amy S Clark
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert K Doot
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, and
| | - Daniel A Pryma
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, and
| | - Sean D Carlin
- Division of Nuclear Medicine Imaging and Therapy, Department of Radiology, and
| |
Collapse
|
164
|
Moreno T, Monterde B, González-Silva L, Betancor-Fernández I, Revilla C, Agraz-Doblas A, Freire J, Isidro P, Quevedo L, Blanco R, Montes-Moreno S, Cereceda L, Astudillo A, Casar B, Crespo P, Morales Torres C, Scaffidi P, Gómez-Román J, Salido E, Varela I. ARID2 deficiency promotes tumor progression and is associated with higher sensitivity to chemotherapy in lung cancer. Oncogene 2021; 40:2923-2935. [PMID: 33742126 PMCID: PMC7610680 DOI: 10.1038/s41388-021-01748-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/23/2021] [Accepted: 03/04/2021] [Indexed: 12/26/2022]
Abstract
The survival rate in lung cancer remains stubbornly low and there is an urgent need for the identification of new therapeutic targets. In the last decade, several members of the SWI/SNF chromatin remodeling complexes have been described altered in different tumor types. Nevertheless, the precise mechanisms of their impact on cancer progression, as well as the application of this knowledge to cancer patient management are largely unknown. In this study, we performed targeted sequencing of a cohort of lung cancer patients on genes involved in chromatin structure. In addition, we studied at the protein level the expression of these genes in cancer samples and performed functional experiments to identify the molecular mechanisms linking alterations of chromatin remodeling genes and tumor development. Remarkably, we found that 20% of lung cancer patients show ARID2 protein loss, partially explained by the presence of ARID2 mutations. In addition, we showed that ARID2 deficiency provokes profound chromatin structural changes altering cell transcriptional programs, which bolsters the proliferative and metastatic potential of the cells both in vitro and in vivo. Moreover, we demonstrated that ARID2 deficiency impairs DNA repair, enhancing the sensitivity of the cells to DNA-damaging agents. Our findings support that ARID2 is a bona fide tumor suppressor gene in lung cancer that may be exploited therapeutically.
Collapse
Affiliation(s)
- Thaidy Moreno
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | - Beatriz Monterde
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | - Laura González-Silva
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | - Isabel Betancor-Fernández
- Departamento de Patología, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Tenerife, Spain
| | - Carlos Revilla
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | - Antonio Agraz-Doblas
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | - Javier Freire
- Servicio de Anatomía Patológica y Biobanco Valdecilla, HUMV/IDIVAL, Santander, Spain
| | - Pablo Isidro
- Biobanco del Principado de Asturias (BBPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Laura Quevedo
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | - Rosa Blanco
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
| | | | - Laura Cereceda
- Servicio de Anatomía Patológica y Biobanco Valdecilla, HUMV/IDIVAL, Santander, Spain
| | - Aurora Astudillo
- Biobanco del Principado de Asturias (BBPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Piero Crespo
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Paola Scaffidi
- Cancer Epigenetics Laboratory, The Francis Crick Institute, London, UK
| | - Javier Gómez-Román
- Servicio de Anatomía Patológica y Biobanco Valdecilla, HUMV/IDIVAL, Santander, Spain
| | - Eduardo Salido
- Departamento de Patología, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Tenerife, Spain
| | - Ignacio Varela
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-CSIC, Santander, Spain.
| |
Collapse
|
165
|
Filho OM, Stover DG, Asad S, Ansell PJ, Watson M, Loibl S, Geyer CE, Bae J, Collier K, Cherian M, O’Shaughnessy J, Untch M, Rugo HS, Huober JB, Golshan M, Sikov WM, von Minckwitz G, Rastogi P, Maag D, Wolmark N, Denkert C, Symmans WF. Association of Immunophenotype With Pathologic Complete Response to Neoadjuvant Chemotherapy for Triple-Negative Breast Cancer: A Secondary Analysis of the BrighTNess Phase 3 Randomized Clinical Trial. JAMA Oncol 2021; 7:603-608. [PMID: 33599688 PMCID: PMC7893540 DOI: 10.1001/jamaoncol.2020.7310] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/07/2020] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Adding carboplatin to standard neoadjuvant chemotherapy (NAC) in triple-negative breast cancer (TNBC) likely benefits a subset of patients; however, determinants of benefit are poorly understood. OBJECTIVE To define the association of molecular subtype, tumor proliferation, and immunophenotype with benefit of carboplatin added to NAC for patients with stages II to III TNBC. DESIGN, SETTING, AND PARTICIPANTS This was a prespecified secondary analysis of a phase 3, double-blind, randomized clinical trial (BrighTNess) that enrolled 634 women across 145 centers in 15 countries. Women with clinical stages II to III TNBC who had undergone pretreatment biopsy were eligible to participate. Whole transcriptome RNA sequencing was performed on the biopsy specimens. The prespecified end point was association of pathologic complete response (pCR) with gene expression-based molecular subtype, with secondary end points investigating established signatures (proliferation, immune) and exploratory analyses of immunophenotype. Data were collected from April 2014 to March 2016. The study analyses were performed from January 2018 to March 2019. INTERVENTIONS Neoadjuvant chemotherapy with paclitaxel followed by doxorubicin and cyclophosphamide, or this same regimen with carboplatin or carboplatin plus veliparib. MAIN OUTCOMES AND MEASURES Association of gene expression-based molecular subtype (PAM50 and TNBC subtypes) with pCR. RESULTS Of the 634 women (median age, 51 [range, 22-78] years) enrolled in BrighTNess, 482 (76%) patients had evaluable RNA sequencing data, with similar baseline characteristics relative to the overall intention-to-treat population. Pathologic complete response was significantly more frequent in PAM50 basal-like vs nonbasal-like cancers overall (202 of 386 [52.3%] vs 34 of 96 [35.4%]; P = .003). Carboplatin benefit was not significantly different in basal-like vs nonbasal-like subgroups (P = .80 for interaction). In multivariable analysis, proliferation (hazard ratio, 0.36; 95% CI, 0.21-0.61; P < .001) and immune (hazard ratio, 0.62; 95% CI, 0.49-0.79; P < .001) signatures were independently associated with pCR. Tumors above the median for proliferation and immune signatures had the highest pCR rate (84 of 125; 67%), while those below the median for both signatures had the lowest pCR rate (42 of 125; 34%). Exploratory gene expression immune analyses suggested that tumors with higher inferred CD8+ T-cell infiltration may receive greater benefit with addition of carboplatin. CONCLUSIONS AND RELEVANCE In this secondary analysis of a randomized clinical trial, triple-negative breast cancer subtyping revealed high pCR rates in basal-like and immunomodulatory subsets. Analysis of biological processes related to basal-like and immunomodulatory phenotypes identified tumor cell proliferation and immune scores as independent factors associated with achieving pCR; the benefit of carboplatin on pCR was seen across all molecular subtypes. Further validation of immunophenotype with existing biomarkers may help to escalate or de-escalate therapy for patients with TNBC. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02032277.
Collapse
Affiliation(s)
- Otto Metzger Filho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Daniel G. Stover
- Department of Medicine, The Ohio State University College of Medicine, Columbus
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus
| | - Sarah Asad
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus
| | | | | | | | - Charles E. Geyer
- Massey Cancer Center, Virginia Commonwealth University, Richmond
- Now with Houston Methodist Cancer Center, Houston, Texas
| | - Junu Bae
- Department of Medicine, The Ohio State University College of Medicine, Columbus
| | - Katharine Collier
- Department of Medicine, The Ohio State University College of Medicine, Columbus
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus
| | - Mathew Cherian
- Department of Medicine, The Ohio State University College of Medicine, Columbus
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus
- AbbVie, Inc, North Chicago, Illinois
| | | | | | | | - Jens B. Huober
- University Medical Center Ulm, Ulm, Germany
- Now with Department of Interdisciplinary Medical Services, Breast Center, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Mehra Golshan
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts
- Now with Yale Cancer Center, New Haven, Connecticut
| | | | | | - Priya Rastogi
- University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, Pennsylvania
| | | | | | - Carsten Denkert
- Institute of Physiology and Pathophysiology, Department of Medicine, Philipps-University Marburg and University Hospital of Giessen and Marburg, Marburg, Germany
| | | |
Collapse
|
166
|
The Affinity of Carboplatin to B-Vitamins and Nucleobases. Int J Mol Sci 2021; 22:ijms22073634. [PMID: 33807309 PMCID: PMC8037198 DOI: 10.3390/ijms22073634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 11/17/2022] Open
Abstract
Platinum compounds have found wide application in the treatment of various types of cancer and carboplatin is one of the main platinum-based drugs used as antitumor agents. The anticancer activity of carboplatin arises from interacting with DNA and inducing programmed cell death. However, such interactions may occur with other chemical compounds, such as vitamins containing aromatic rings with lone-pair orbitals, which reduces the anti-cancer effect of carboplatin. The most important aspect of the conducted research was related to the evaluation of carboplatin affinity to vitamins from the B group and the potential impact of such interactions on the reduction of therapeutic capabilities of carboplatin in anticancer therapy. Realized computations, including estimation of Gibbs Free Energies, allowed for the identification of the most reactive molecule, namely vitamin B6 (pyridoxal phosphate). In this case, the computational estimations indicating carboplatin reactivity were confirmed by spectrophotometric measurements.
Collapse
|
167
|
Ouma LO, Grayling MJ, Zheng H, Wason J. Treatment allocation strategies for umbrella trials in the presence of multiple biomarkers: A comparison of methods. Pharm Stat 2021; 20:990-1001. [PMID: 33759353 PMCID: PMC7612600 DOI: 10.1002/pst.2119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/01/2021] [Accepted: 03/13/2021] [Indexed: 12/29/2022]
Abstract
Umbrella trials are an innovative trial design where different treatments are matched with subtypes of a disease, with the matching typically based on a set of biomarkers. Consequently, when patients can be positive for more than one biomarker, they may be eligible for multiple treatment arms. In practice, different approaches could be applied to allocate patients who are positive for multiple biomarkers to treatments. However, to date there has been little exploration of how these approaches compare statistically. We conduct a simulation study to compare five approaches to handling treatment allocation in the presence of multiple biomarkers – equal randomisation; randomisation with fixed probability of allocation to control; Bayesian adaptive randomisation (BAR); constrained randomisation; and hierarchy of biomarkers. We evaluate these approaches under different scenarios in the context of a hypothetical phase II biomarker-guided umbrella trial. We define the pairings representing the pre-trial expectations on efficacy as linked pairs, and the other biomarker-treatment pairings as unlinked. The hierarchy and BAR approaches have the highest power to detect a treatment-biomarker linked interaction. However, the hierarchy procedure performs poorly if the pre-specified treatment-biomarker pairings are incorrect. The BAR method allocates a higher proportion of patients who are positive for multiple biomarkers to promising treatments when an unlinked interaction is present. In most scenarios, the constrained randomisation approach best balances allocation to all treatment arms. Pre-specification of an approach to deal with treatment allocation in the presence of multiple biomarkers is important, especially when overlapping subgroups are likely.
Collapse
Affiliation(s)
- Luke Ondijo Ouma
- Biostatistics Research Group, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Michael J Grayling
- Biostatistics Research Group, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Haiyan Zheng
- Biostatistics Research Group, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James Wason
- Biostatistics Research Group, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.,MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
168
|
Abstract
PURPOSE OF REVIEW Platinum compounds are used in the treatment of various types of cancer. Here, we review the current role of cisplatin and carboplatin in the treatment of early stage and advanced triple-negative breast cancer (TNBC), and the use of biomarkers in predicting response to platinum therapy. RECENT FINDINGS Addition of carboplatin to a neoadjuvant chemotherapy regimen can result in improvement in the pathological complete response rates. The long-term benefit of the addition of carboplatin to standard chemotherapy regimens remains unproven. Single-agent platinum is an option in the treatment of advanced breast cancer. BRCA1/2 mutations predicted benefit from platinums in advanced, but not early stage breast cancer. There are yet no biomarkers to predict response to platinum in sporadic TNBC. Platinum compounds are an option in the treatment of TNBC. Identification of biomarkers to select tumors most likely to derive benefit from these agents is still needed. Ongoing trials are exploring the role of platinum in the adjuvant setting and in combination with other agents, including immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Filipa Lynce
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave YC-1275, Boston, MA, 02215, USA.
| | - Raquel Nunes
- Medical Oncology, Johns Hopkins Sidney Kimmel Cancer Center at Sibley Memorial Hospital, Building B, First Floor, 5255 Loughboro Rd, NW, Washington, DC, 20016, USA
| |
Collapse
|
169
|
Nitecki R, Melamed A, Gockley AA, Floyd J, Krause KJ, Coleman RL, Matulonis UA, Giordano SH, Lu KH, Rauh-Hain JA. Incidence of myelodysplastic syndrome and acute myeloid leukemia in patients receiving poly-ADP ribose polymerase inhibitors for the treatment of solid tumors: A meta-analysis of randomized trials. Gynecol Oncol 2021; 161:653-659. [PMID: 33736856 DOI: 10.1016/j.ygyno.2021.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinical trials demonstrated that PARPi (poly [adenosine diphosphate-ribose]-ADP polymerase inhibitor) therapy is effective in solid tumors. However, long term effects such as therapy-related myelodysplastic syndrome or acute myeloid leukemia (MDS/AML) are poorly described. We sought to quantify whether PARPi therapy is associated with the development of MDS/AML. METHODS Medline, Embase, and Cochrane databases were searched (inception to January 6, 2020) and phase 2 and 3 clinical trials that randomized patients with solid tumors to a PARPi or control therapy were included. The PRISMA guidelines were used to extract data independently by multiple authors. We extracted person-time and number of cases of MDS/AML in the PARPi and control arms of each study and pooled results with a random-effects Poisson regression model. The pooled incidence rate ratio (IRR) for MDS/AML among patients randomized to PARPi therapy was compared to those randomized to a control. RESULTS We identified 14 studies that included 5739 patients. Accounting for intra-study clustering, the risk of MDS/AML was similar in patients who were randomly assigned to receive PARPi compared to controls (IRR 1.32, 95% confidence interval [CI] 0.78-2.26). In the front-line setting, PARPi therapy was associated with developing MDS/AML (IRR 5.43, 95% CI 1.51-19.60). Among patients treated for recurrence, however, the risk of MDS/AML appeared to be similar among patients randomized to PARPi or control treatment. Among studies that included only patients with a BRCA mutation, the risk of MDS/AML was similar in both treatment groups (IRR 0.83, 95% CI 0.45-1.53), but PARPi therapy was associated with MDS/AML in studies with an unrestricted population (IRR 2.43, 95% CI 1.17-5.06). CONCLUSION The pooled overall effect was not statistically significant. However, treatment with PARPi was associated with a statistically significant increase in the incidence of MDS/AML among patients receiving front-line cancer therapy and those with limited prior exposure to chemotherapy.
Collapse
Affiliation(s)
- Roni Nitecki
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Alexander Melamed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
| | - Allison A Gockley
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
| | - Jessica Floyd
- Department of Obstetrics and Gynecology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, United States of America
| | - Kate J Krause
- Research Medical Library, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Robert L Coleman
- US Oncology Research, The Woodlands, TX, United States of America
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States of America
| | - Sharon H Giordano
- Departments of Health Services Research and Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - J Alejandro Rauh-Hain
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.
| |
Collapse
|
170
|
Combining poly(ADP-ribose) polymerase inhibitors and immune checkpoint inhibitors in breast cancer: rationale and preliminary clinical results. Curr Opin Oncol 2021; 32:585-593. [PMID: 32852311 DOI: 10.1097/cco.0000000000000680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Recently, both immune checkpoint inhibitors and poly(ADP-ribose) polymerase inhibitors have demonstrated clinical benefit in some subsets of HER2-negative breast cancer patients. A biological rationale exists supporting a potential synergism between these compounds, which may further increase their antitumor activity in the clinic. RECENT FINDINGS PARP inhibitors were shown to activate type I interferon pathway, thus eliciting local and general immune response, while inducing programmed cell death-ligand 1 (PD-L1) up-regulation. In addition, the DNA damages created by PARP inhibition may increase tumor mutational burden and neo-antigens, thereby favoring efficacy of immune checkpoint inhibitors. Accordingly, clinical trials combining PARP inhibitors and agents targeting the PD-1/PD-L1 axis have been initiated in breast cancer in both advanced and early stages, enrolling patients with germline BRCA1/2 mutation, homologous recombination deficiency and/or with triple negative phenotype. Preliminary safety and efficacy results are encouraging, but it is still unclear whether the combination adds benefit compared with each therapeutic administered as single agent. SUMMARY Although a strong rationale exists to support the combination of PARP inhibitors with immune checkpoint inhibitors, future clinical trials will have to demonstrate whether it improves outcome and to identify which patients are the most likely to benefit from.
Collapse
|
171
|
Esposito CL, Quintavalle C, Ingenito F, Rotoli D, Roscigno G, Nuzzo S, Thomas R, Catuogno S, de Franciscis V, Condorelli G. Identification of a novel RNA aptamer that selectively targets breast cancer exosomes. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:982-994. [PMID: 33614245 PMCID: PMC7868932 DOI: 10.1016/j.omtn.2021.01.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is a leading cause of cancer mortality in women. Despite advances in its management, the identification of new options for early-stage diagnosis and therapy of this tumor still represents a crucial challenge. Increasing evidence indicates that extracellular vesicles called exosomes may have great potential as early diagnostic biomarkers and regulators of many cancers, including breast cancer. Therefore, exploiting molecules able to selectively recognize them is of great interest. Here, we developed a novel differential SELEX strategy, called Exo-SELEX, to isolate nucleic acid aptamers against intact exosomes derived from primary breast cancer cells. Among the obtained sequences, we optimized a high-affinity aptamer (ex-50.T) able to specifically recognize exosomes from breast cancer cells or patient serum samples. Furthermore, we demonstrated that the ex.50.T is a functional inhibitor of exosome cellular uptake and antagonizes cancer exosome-induced cell migration in vitro. This molecule provides an innovative tool for the specific exosome detection and the development of new therapeutic approaches for breast cancer.
Collapse
Affiliation(s)
- Carla Lucia Esposito
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Cristina Quintavalle
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Francesco Ingenito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II,” 80100 Naples, Italy
| | - Deborah Rotoli
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Giuseppina Roscigno
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II,” 80100 Naples, Italy
| | | | | | - Silvia Catuogno
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Vittorio de Franciscis
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Gerolama Condorelli
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II,” 80100 Naples, Italy
| |
Collapse
|
172
|
Obeng-Gyasi S, Coles CE, Jones J, Sacks R, Lightowlers S, Bliss JM, Brunt AM, Haviland JS, Kirby AM, Kalinsky K. When the World Throws You a Curve Ball: Lessons Learned in Breast Cancer Management. Am Soc Clin Oncol Educ Book 2021; 41:1-11. [PMID: 33956493 DOI: 10.1200/edbk_320691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the care of patients with operable breast cancer, there has been a shift toward increasing use of neoadjuvant therapy. There are benefits to neoadjuvant therapy, such as monitoring for response, as well as an increased rate of breast conservation and reduction of potential morbidity associated with breast surgery, including axillary management. Among patients with highly proliferative tumors, such as HER2-positive or triple-negative breast cancer, those with residual disease are at higher risk of recurrence, which informs the recommended systemic therapy in the adjuvant setting. For instance, in patients with residual disease after neoadjuvant chemotherapy and HER2-targeted therapy, there is a role for adjuvant trastuzumab emtansine for those with residual disease at the time of surgery. The same holds true regarding the role of adjuvant capecitabine in patients with residual disease after neoadjuvant chemotherapy. With the added complexities of treating patients in the era of the COVID-19 outbreak, additional considerations are critical, including initiation of surgery within an appropriate time from completion of neoadjuvant therapy. National consensus guidelines on time to surgery must be developed to improve measurement and comparison across systems. In addition, there is emerging radiation treatment management research addressing a number of factors, including hypofractionation, role of proton beam therapy, safe omission of radiotherapy, and preoperative radiotherapy with or without drug combination. In this article, the multidisciplinary approach of treating patients with operable breast cancer is highlighted, with updates and future considerations described.
Collapse
Affiliation(s)
- Samilia Obeng-Gyasi
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, OH
| | - Charlotte E Coles
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Jade Jones
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Ruth Sacks
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Sara Lightowlers
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Judith M Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - A Murray Brunt
- School of Medicine, University of Keele, Keele, United Kingdom
| | - Joanne S Haviland
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Anna M Kirby
- Department of Radiotherapy, Royal Marsden National Health Service Foundation Trust and The Institute of Cancer Research, London, United Kingdom
| | - Kevin Kalinsky
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
173
|
Du L, Yau C, Brown-Swigart L, Gould R, Krings G, Hirst GL, Bedrosian I, Layman RM, Carter JM, Klein M, Venters S, Shad S, van der Noordaa M, Chien AJ, Haddad T, Isaacs C, Pusztai L, Albain K, Nanda R, Tripathy D, Liu MC, Boughey J, Schwab R, Hylton N, DeMichele A, Perlmutter J, Yee D, Berry D, Van't Veer L, Valero V, Esserman LJ, Symmans WF. Predicted sensitivity to endocrine therapy for stage II-III hormone receptor-positive and HER2-negative (HR+/HER2-) breast cancer before chemo-endocrine therapy. Ann Oncol 2021; 32:642-651. [PMID: 33617937 DOI: 10.1016/j.annonc.2021.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/07/2021] [Accepted: 02/13/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND We proposed that a test for sensitivity to the adjuvant endocrine therapy component of treatment for patients with stage II-III breast cancer (SET2,3) should measure transcription related to estrogen and progesterone receptors (SETER/PR index) adjusted for a baseline prognostic index (BPI) combining clinical tumor and nodal stage with molecular subtype by RNA4 (ESR1, PGR, ERBB2, and AURKA). PATIENTS AND METHODS Patients with clinically high-risk, hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-negative (HR+/HER2-) breast cancer received neoadjuvant taxane-anthracycline chemotherapy, surgery with measurement of residual cancer burden (RCB), and then adjuvant endocrine therapy. SET2,3 was measured from pre-treatment tumor biopsies, evaluated first in an MD Anderson Cancer Center (MDACC) cohort (n = 307, 11 years' follow-up, U133A microarrays), cut point was determined, and then independent, blinded evaluation was carried out in the I-SPY2 trial (n = 268, high-risk MammaPrint result, 3.8 years' follow-up, Agilent-44K microarrays, NCI Clinical Trials ID: NCT01042379). Primary outcome measure was distant relapse-free survival. Multivariate Cox regression models tested prognostic independence of SET2,3 relative to RCB and other molecular prognostic signatures, and whether other prognostic signatures could substitute for SETER/PR or RNA4 components of SET2,3. RESULTS SET2,3 added independent prognostic information to RCB in the MDACC cohort: SET2,3 [hazard ratio (HR) 0.23, P = 0.004] and RCB (HR 1.77, P < 0.001); and the I-SPY2 trial: SET2,3 (HR 0.27, P = 0.031) and RCB (HR 1.68, P = 0.008). SET2,3 provided similar prognostic information irrespective of whether RCB-II or RCB-III after chemotherapy, and in both luminal subtypes. Conversely, RCB was most strongly prognostic in cancers with low SET2,3 status (MDACC P < 0.001, I-SPY2 P < 0.001). Other molecular signatures were not independently prognostic; they could effectively substitute for RNA4 subtype within the BPI component of SET2,3, but they could not effectively substitute for SETER/PR index. CONCLUSIONS SET2,3 added independent prognostic information to chemotherapy response (RCB) and baseline prognostic score or subtype. Approximately 40% of patients with clinically high-risk HR+/HER2- disease had high SET2,3 and could be considered for clinical trials of neoadjuvant endocrine-based treatment.
Collapse
Affiliation(s)
- L Du
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Yau
- Department of Surgery, University of California, San Francisco, USA
| | - L Brown-Swigart
- Department of Pathology, University of California, San Francisco, USA
| | - R Gould
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - G Krings
- Department of Pathology, University of California, San Francisco, USA
| | - G L Hirst
- Department of Surgery, University of California, San Francisco, USA
| | - I Bedrosian
- Department of Breast Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - R M Layman
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J M Carter
- Department of Pathology, Mayo Clinic, Rochester, USA
| | - M Klein
- Department of Pathology, University of Minnesota, Minneapolis, USA
| | - S Venters
- Department of Surgery, University of California, San Francisco, USA
| | - S Shad
- Department of Surgery, University of California, San Francisco, USA
| | | | - A J Chien
- Department of Medicine, University of California, San Francisco, USA
| | - T Haddad
- Department of Medicine, Mayo Clinic, Rochester, USA
| | - C Isaacs
- Department of Medicine, Georgetown University, Washington, USA
| | - L Pusztai
- Department of Medicine, Yale University School of Medicine, New Haven, USA
| | - K Albain
- Department of Medicine, Loyola University, Chicago, USA
| | - R Nanda
- Department of Medicine, University of Chicago, Chicago, USA
| | - D Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M C Liu
- Department of Medicine, Mayo Clinic, Rochester, USA
| | - J Boughey
- Department of Surgery, Mayo Clinic, Rochester, USA
| | - R Schwab
- Department of Medicine, University of California, San Diego, USA
| | - N Hylton
- Department of Radiology, University of California, San Francisco, USA
| | - A DeMichele
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, San Philadelphia, USA
| | | | - D Yee
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - D Berry
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L Van't Veer
- Department of Pathology, University of California, San Francisco, USA
| | - V Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L J Esserman
- Department of Surgery, University of California, San Francisco, USA
| | - W F Symmans
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Pathology, The University of Texas MD Anderson Cancer Center, San Francisco, USA.
| |
Collapse
|
174
|
Fleisher B, Lezeau J, Werkman C, Jacobs B, Ait-Oudhia S. In vitro to Clinical Translation of Combinatorial Effects of Doxorubicin and Abemaciclib in Rb-Positive Triple Negative Breast Cancer: A Systems-Based Pharmacokinetic/Pharmacodynamic Modeling Approach. BREAST CANCER-TARGETS AND THERAPY 2021; 13:87-105. [PMID: 33628047 PMCID: PMC7899308 DOI: 10.2147/bctt.s292161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/19/2021] [Indexed: 11/23/2022]
Abstract
Background Doxorubicin (DOX) and its pegylated liposomal formulation (L_DOX) are the standard of care for triple-negative breast cancer (TNBC). However, resistance to DOX often occurs, motivating the search for alternative treatment approaches. The retinoblastoma protein (Rb) is a potential pharmacological target for TNBC treatment since its expression has been associated with resistance to DOX-based therapy. Methods DOX (0.01–20 μM) combination with abemaciclib (ABE, 1–6 μM) was evaluated over 72 hours on Rb-positive (MDA-MB-231) and Rb-negative (MDA-MB-468) TNBC cells. Combination indices (CI) for DOX+ABE were calculated using Compusyn software. The TNBC cell viability time-course and fold-change from the control of phosphorylated-Rb (pRb) protein expression were measured with CCK8-kit and enzyme-linked immunosorbent assay. A cell-based pharmacodynamic (PD) model was developed, where pRb protein dynamics drove cell viability response. Clinical pharmacokinetic (PK) models for DOX, L_DOX, and ABE were developed using data extracted from the literature. After scaling cancer cell growth to clinical TNBC tumor growth, the time-to-tumor progression (TTP) was predicted for human dosing regimens of DOX, ABE, and DOX+ABE. Results DOX and ABE combinations were synergistic (CI<1) in MDA-MB-231 and antagonistic (CI>1) in MDA-MB-468. The maximum inhibitory effects (Imax) for both drugs were set to one. The drug concentrations producing 50% of Imax for DOX and ABE were 0.565 and 2.31 μM (MDA-MB-231) and 0.121 and 1.61 μM (MDA-MB-468). The first-orders rate constants of abemaciclib absorption (ka) and doxorubicin release from L_DOX (kRel) were estimated at 0.31 and 0.013 h−1. Their linear clearances were 21.7 (ABE) and 32.1 L/h (DOX). The estimated TTP for intravenous DOX (75 mg/m2 every 21 days), intravenous L_DOX (50 mg/m2 every 28 days), and oral ABE (200 mg twice a day) were 125, 31.2, and 8.6 days shorter than drug-free control. The TTP for DOX+ABE and L_DOX+ABE were 312 days and 47.5 days shorter than control, both larger than single-agent DOX, suggesting improved activity with the DOX+ABE combination. Conclusion The developed translational systems-based PK/PD model provides an in vitro-to-clinic modeling platform for DOX+ABE in TNBC. Although model-based simulations suggest improved outcomes with combination over monotherapy, tumor relapse was not prevented with the combination. Hence, DOX+ABE may not be an effective treatment combination for TNBC.
Collapse
Affiliation(s)
- Brett Fleisher
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Jovin Lezeau
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Carolin Werkman
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Brehanna Jacobs
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Sihem Ait-Oudhia
- Quantitative Pharmacology and Pharmacometrics (QP2), Merck & Co, Inc, Kenilworth, New Jersey, USA
| |
Collapse
|
175
|
DNA damage response inhibitors: An avenue for TNBC treatment. Biochim Biophys Acta Rev Cancer 2021; 1875:188521. [PMID: 33556453 DOI: 10.1016/j.bbcan.2021.188521] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/30/2021] [Accepted: 01/30/2021] [Indexed: 01/13/2023]
Abstract
The DNA damage response (DDR) is critical for the maintenance of genomic stability by sensing DNA damage, regulating cell cycle and initiating DNA repair. Drugs targeting DDR pathways have been increasingly exploited in treating various tumors. Triple negative breast cancer (TNBC) is a highly heterogeneous and aggressive tumor with constitutive activation of oncogenes, inducing replication stress and DNA damage, which require the DDR for survival. In addition, emerging studies have demonstrated that TNBC harbors aberrant genetic alterations in DDR pathways, such as a high frequency of p53 dysfunction and BRCA1/2 mutations. DDR alterations force TNBC to rely on the existing DDR pathways for survival, and make TNBC particularly sensitive to specific DDR inhibitors, such as high sensitivity of TNBC with BRCA1/2 mutations to PARP inhibitors. This review first and comprehensively covers the current status of the development of DDR inhibitors and discusses the mechanism of targeting the DDR in TNBC. Preclinical and clinical studies on inhibitors of the ATR-CHK1-WEE1 pathway and PARP inhibitors, the most studied inhibitors, and some other DDR inhibitors as monotherapy or combination therapy in TNBC are summarized. We also highlight the possible predictive biomarkers for these DDR inhibitors and their potential combination strategies with chemotherapy, radiotherapy or other targeted agents to optimize the efficacy of DDR inhibitors in TNBC treatment. In conclusion, this review discussed the recent considerations related to the use of DDR inhibitors for TNBC and provides a perspective to address future directions and potential therapeutic strategies for patients with TNBC.
Collapse
|
176
|
Tufano AM, Teplinsky E, Landry CA. Updates in Neoadjuvant Therapy for Triple Negative Breast Cancer. Clin Breast Cancer 2021; 21:1-9. [DOI: 10.1016/j.clbc.2020.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 06/28/2020] [Accepted: 07/01/2020] [Indexed: 11/25/2022]
|
177
|
McCann KE, Hurvitz SA. Innovations in targeted therapies for triple negative breast cancer. Curr Opin Obstet Gynecol 2021; 33:34-47. [PMID: 33093337 DOI: 10.1097/gco.0000000000000671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Triple negative breast cancer (TNBC) is defined by a lack of targets, namely hormone receptor (HR) expression and human epidermal growth factor receptor 2 amplification. Cytotoxic chemotherapy remains the mainstay of treatment. Though TNBC constitutes approximately 10-15% of breast cancer, it is disproportionally lethal, but it is hoped that outcomes will improve as targetable oncogenic drivers are identified. RECENT FINDINGS Translational work in TNBC has focused on subsets defined by defects in homologous recombination repair, immune cell infiltration, or programmed death ligand receptor 1 expression, an over-active phosphoinositide-3 kinase pathway, or expression of androgen receptors. Though not specific to TNBC, the novel cell surface antigen trophoblast antigen 2 has also been identified and successfully targeted. This work has led to Food and Drug Administration approvals for small molecule poly-ADP-ribosyl polymerase inhibitors in patients with deleterious germline mutations in BRCA1 or BRCA2, the combination of nab-paclitaxel with immune checkpoint inhibitor antibodies in the first-line metastatic setting for programmed death ligand receptor 1+ TNBC, and use of the antibody-drug conjugate sacituzumab govitecan in the later-line metastatic setting. SUMMARY Identification of targetable oncogenic drivers in TNBC is an area of intense cancer biology research, hopefully translating to new therapies and improved outcomes.
Collapse
Affiliation(s)
- Kelly E McCann
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
178
|
PARP Inhibitors in Triple-Negative Breast Cancer Including Those With BRCA Mutations. ACTA ACUST UNITED AC 2021; 27:67-75. [PMID: 33475295 DOI: 10.1097/ppo.0000000000000499] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
ABSTRACT Poly(ADP-ribose) polymerase (PARP) is involved in single-strand DNA break base excision repair. PARP inhibition causes synthetic lethality in breast cancers associated with germline BRCA1 and BRCA2 mutations and is routinely used in clinical practice for metastatic breast cancer. Breast cancers with homologous recombination deficiency or BRCAness, most commonly triple-negative breast cancers, may also benefit. Currently, PARP inhibitor use for triple-negative breast cancer with wild-type BRCA does not have definitive efficacy; however, this is an area of active research. Further clinical and translational data may identify additional patient populations that will benefit from PARP inhibitor therapy.
Collapse
|
179
|
Abstract
ABSTRACT Triple-negative breast cancer (TNBC) is an aggressive subtype of mammary carcinoma. A subset of TNBC is immune activated, suggesting that immunotherapy may be a viable treatment strategy. Phase III clinical trials have shown that atezolizumab or pembrolizumab is well-tolerated in combination with chemotherapy, with progression-free survival benefit in metastatic programmed death ligand-1 (PD-L1)-positive TNBC patients treated first line. Based on IMpassion130, the combination of atezolizumab and nab-paclitaxel is now considered a standard of care for the treatment of PD-L1-positive advanced TNBC. In early TNBC, pembrolizumab and atezolizumab have been tested in combination with standard neoadjuvant chemotherapy, resulting in a higher complete pathologic response rate than standard neoadjuvant chemotherapy alone, regardless of disease PD-L1 status. These findings establish proof of principle for immunotherapy in both early and advanced TNBC. High priorities for the field include developing more active immunotherapy combination regimens and more refined biomarkers that optimally identify patients most likely to benefit from immunotherapy.
Collapse
|
180
|
Frequency and Outcomes of New Suspicious Lesions on Breast MRI in the Setting of Neoadjuvant Therapy. AJR Am J Roentgenol 2021; 216:633-639. [PMID: 33439044 DOI: 10.2214/ajr.20.22979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE. The purpose of this article was to determine the frequency and outcomes of new suspicious findings on breast MRI after initiation of neoadjuvant therapy. MATERIALS AND METHODS. A retrospective database review identified all breast MRI examinations performed to assess response to neoadjuvant therapy between 2010 and 2018. Cases with new suspicious lesions assessed as BI-RADS 4 or 5 and found after the initiation of neoadjuvant treatment were included. Cases with no pretreatment MRI, cases in which the suspicious lesion was present on the baseline MRI but remained suspicious, and cases with insufficient follow-up were excluded. Radiologic, pathologic, and surgical reports were reviewed. Malignant outcomes were determined by pathologic examination. Benignity was established by pathologic examination, follow-up imaging, or both. A total of 419 breast MRI examinations in 297 women were performed to assess response to neoadjuvant therapy. After exclusions, 23 MRI examinations (5.5%) with new suspicious findings, all assessed as BI-RADS 4, comprised the final cohort. RESULTS. Of the 23 lesions, 13 new suspicious findings (56.5%) were contralateral to the known malignancy, nine (39.1%) were ipsilateral, and one (4.3%) involved the bilateral breasts. Lesion types included mass (16, 69.6%), nonmass enhancement (5, 21.7%) and focus (2, 8.7%). None of the new suspicious findings were malignant. CONCLUSION. New suspicious findings occurred in 5.5% of breast MRI examinations performed to monitor response to neoadjuvant therapy, and none of these new lesions were malignant. Our findings suggest that new lesions that arise in the setting of neoadjuvant therapy are highly unlikely to represent a new site of malignancy, particularly if the index malignancy shows treatment response. Larger studies are needed to confirm whether biopsy may be safely averted in this scenario.
Collapse
|
181
|
Chen AP, Kummar S, Moore N, Rubinstein LV, Zhao Y, Williams PM, Palmisano A, Sims D, O'Sullivan Coyne G, Rosenberger CL, Simpson M, Raghav KPS, Meric-Bernstam F, Leong S, Waqar S, Foster JC, Konaté MM, Das B, Karlovich C, Lih CJ, Polley E, Simon R, Li MC, Piekarz R, Doroshow JH. Molecular Profiling-Based Assignment of Cancer Therapy (NCI-MPACT): A Randomized Multicenter Phase II Trial. JCO Precis Oncol 2021; 5:PO.20.00372. [PMID: 33928209 PMCID: PMC8078898 DOI: 10.1200/po.20.00372] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/10/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
This trial assessed the utility of applying tumor DNA sequencing to treatment selection for patients with advanced, refractory cancer and somatic mutations in one of four signaling pathways by comparing the efficacy of four study regimens that were either matched to the patient's aberrant pathway (experimental arm) or not matched to that pathway (control arm). MATERIALS AND METHODS Adult patients with an actionable mutation of interest were randomly assigned 2:1 to receive either (1) a study regimen identified to target the aberrant pathway found in their tumor (veliparib with temozolomide or adavosertib with carboplatin [DNA repair pathway], everolimus [PI3K pathway], or trametinib [RAS/RAF/MEK pathway]), or (2) one of the same four regimens, but chosen from among those not targeting that pathway. RESULTS Among 49 patients treated in the experimental arm, the objective response rate was 2% (95% CI, 0% to 10.9%). One of 20 patients (5%) in the experimental trametinib cohort had a partial response. There were no responses in the other cohorts. Although patients and physicians were blinded to the sequencing and random assignment results, a higher pretreatment dropout rate was observed in the control arm (22%) compared with the experimental arm (6%; P = .038), suggesting that some patients may have had prior tumor mutation profiling performed that led to a lack of participation in the control arm. CONCLUSION Further investigation, better annotation of predictive biomarkers, and the development of more effective agents are necessary to inform treatment decisions in an era of precision cancer medicine. Increasing prevalence of tumor mutation profiling and preference for targeted therapy make it difficult to use a randomized phase II design to evaluate targeted therapy efficacy in an advanced disease setting.
Collapse
Affiliation(s)
- Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shivaani Kummar
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR
| | - Nancy Moore
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Yingdong Zhao
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - P. Mickey Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Alida Palmisano
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- General Dynamics Information Technology (GDIT), Falls Church, VA
| | - David Sims
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Mel Simpson
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kanwal P. S. Raghav
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Funda Meric-Bernstam
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Saiama Waqar
- Department of Medical Oncology, Washington University School of Medicine, St Louis, MO
| | - Jared C. Foster
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Mariam M. Konaté
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Biswajit Das
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chris Karlovich
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Chih-Jian Lih
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Eric Polley
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Richard Simon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Ming-Chung Li
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Richard Piekarz
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - James H. Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
- Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
182
|
Wu HJ, Chu PY. Recent Discoveries of Macromolecule- and Cell-Based Biomarkers and Therapeutic Implications in Breast Cancer. Int J Mol Sci 2021; 22:ijms22020636. [PMID: 33435254 PMCID: PMC7827149 DOI: 10.3390/ijms22020636] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer type and the leading cause of cancer-related mortality in women worldwide. Breast cancer is fairly heterogeneous and reveals six molecular subtypes: luminal A, luminal B, HER2+, basal-like subtype (ER−, PR−, and HER2−), normal breast-like, and claudin-low. Breast cancer screening and early diagnosis play critical roles in improving therapeutic outcomes and prognosis. Mammography is currently the main commercially available detection method for breast cancer; however, it has numerous limitations. Therefore, reliable noninvasive diagnostic and prognostic biomarkers are required. Biomarkers used in cancer range from macromolecules, such as DNA, RNA, and proteins, to whole cells. Biomarkers for cancer risk, diagnosis, proliferation, metastasis, drug resistance, and prognosis have been identified in breast cancer. In addition, there is currently a greater demand for personalized or precise treatments; moreover, the identification of novel biomarkers to further the development of new drugs is urgently needed. In this review, we summarize and focus on the recent discoveries of promising macromolecules and cell-based biomarkers for the diagnosis and prognosis of breast cancer and provide implications for therapeutic strategies.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua County 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 231, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, No. 542, Sec. 1 Chung-Shan Rd., Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975-611-855; Fax: +886-4-7227-116
| |
Collapse
|
183
|
The Bayesian Design of Adaptive Clinical Trials. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18020530. [PMID: 33435249 PMCID: PMC7826635 DOI: 10.3390/ijerph18020530] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 01/13/2023]
Abstract
This paper presents a brief overview of the recent literature on adaptive design of clinical trials from a Bayesian perspective for statistically not so sophisticated readers. Adaptive designs are attracting a keen interest in several disciplines, from a theoretical viewpoint and also—potentially—from a practical one, and Bayesian adaptive designs, in particular, have raised high expectations in clinical trials. The main conceptual tools are highlighted here, with a mention of several trial designs proposed in the literature that use these methods, including some of the registered Bayesian adaptive trials to this date. This review aims at complementing the existing ones on this topic, pointing at further interesting reading material.
Collapse
|
184
|
Wang C, Li J. Haematologic toxicities with PARP inhibitors in cancer patients: an up-to-date meta-analysis of 29 randomized controlled trials. J Clin Pharm Ther 2021; 46:571-584. [PMID: 33421183 DOI: 10.1111/jcpt.13349] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Poly ADP-ribose polymerase (PARP) inhibitors have emerged as one of the most exciting new treatments for patients with certain types of cancer. Haematologic toxicities are common adverse events (AEs) for all PARP inhibitors. We conducted a meta-analysis to fully investigate the haematologic toxicities of PARP inhibitors in cancer patients. STUDY DESIGN PubMed/Medline and Embase were searched for articles published till September 2020. The relevant randomized controlled trials (RCTs) in cancer patients treated with PARP inhibitors were retrieved, and the systematic evaluation was performed. RESULTS Twenty-nine RCTs and 9247 patients were included. The current meta-analysis suggests that the use of PARP inhibitors significantly increases the risk of all-grade anaemia (RR, 2.32; 95% CI, 1.78-3.01; p < 0.00001), neutropenia (RR, 1.69; 95% CI, 1.38-2.07; p < 0.00001) and thrombocytopenia (RR, 2.54; 95% CI, 1.87-3.45; p < 0.00001). The use of these agents also significantly increased the risk of high-grade anaemia (RR, 3.06; 95% CI, 2.11-4.43; p < 0.00001), neutropenia (RR, 1.66; 95% CI, 1.33-2.07; p < 0.00001) and thrombocytopenia (RR, 2.76; 95% CI, 1.83-4.16; p < 0.00001). Anaemia was the most common haematologic toxicity, and all the five included PARP inhibitors were associated with a significant increased risk of anaemia. Combination treatment may reduce the risk of anaemia and thrombocytopenia compared to those receiving PARP inhibitor monotherapy. WHAT IS NEW AND CONCLUSION The available data suggested that the use of PARP inhibitors was associated with a significantly increased risk of haematologic toxicities.
Collapse
Affiliation(s)
- Chengyuan Wang
- College of Environment and Resource, Chongqing Technology and Business University, Chongqing, P.R.China
| | - Jing Li
- College of Pharmacy, Southwest Minzu University, Sichuan, P.R.China
| |
Collapse
|
185
|
Froeling FEM, Casolino R, Pea A, Biankin AV, Chang DK, on behalf of Precision-Panc. Molecular Subtyping and Precision Medicine for Pancreatic Cancer. J Clin Med 2021; 10:E149. [PMID: 33406790 PMCID: PMC7794969 DOI: 10.3390/jcm10010149] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/13/2020] [Accepted: 12/25/2020] [Indexed: 12/12/2022] Open
Abstract
Substantial progress in recent years has dramatically increased our knowledge of the molecular basis of cancer, revealing new potential therapeutic targets and paving the way for effective personalised medicine for the treatment of many tumour types. However, pancreatic cancer has been lagging behind in this success and continues to be one of the most lethal solid malignancies. Its molecular heterogeneity and the unselected design of the majority of clinical trials to date can in part explain the reason for our failure to make a significant change in the survival outcomes for patients with pancreatic cancer. A changing paradigm in drug development is required to validate the new molecular taxonomy and to rapidly translate preclinical discovery into clinical trials. Here, we review the molecular subtyping of pancreatic cancer, the challenges in identifying effective treatment regimens according to defined low-prevalence molecular subgroups and we illustrate a new model of translational therapeutic development that was established in the U.K. (Precision-Panc) as a potentially effective solution to improve outcomes for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Fieke E. M. Froeling
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Raffaella Casolino
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- Department of Medicine, University and Hospital Trust of Verona of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Antonio Pea
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- Department of Surgery, University and Hospital Trust of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Andrew V. Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
| | - David K. Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; (F.E.M.F.); (R.C.); (A.P.); (A.V.B.)
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow G31 2ER, UK
| | | |
Collapse
|
186
|
Potter DA, Herrera-Ponzanelli CA, Hinojosa D, Castillo R, Hernandez-Cruz I, Arrieta VA, Franklin MJ, Yee D. Recent advances in neoadjuvant therapy for breast cancer. Fac Rev 2021; 10:2. [PMID: 33659921 PMCID: PMC7894264 DOI: 10.12703/r/10-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neoadjuvant trials for early breast cancer have accelerated the identification of novel active agents, enabling streamlined conduct of registration trials with fewer subjects. Measurement of neoadjuvant drug effects has also enabled the identification of patients with high risk of distant recurrence and has justified development of additional adjuvant approaches to improve outcomes. Neoadjuvant evaluation of new drugs was significantly improved by the introduction of pathologic complete response (pCR) rate as a quantitative surrogate endpoint for distant disease-free survival (DDFS) and event free survival (EFS). The neoadjuvant phase 2 platform trial I-SPY 2 simultaneously tests multiple drugs across multiple breast cancer subtypes using Bayesian methods of adaptive randomization for assessment of drug efficacy. In addition to the pCR endpoint, the I-SPY 2 trial has demonstrated that the residual cancer burden (RCB) score measures gradations of tumor response that correlate with DDFS and EFS across treatments and subtypes. For HER2-positive and triple-negative breast cancers that have failed to attain pCR with neoadjuvant chemotherapy (NAC), effective modifications of adjuvant treatment have improved outcomes and changed the standard of care for these subtypes. Neoadjuvant therapy is therefore preferred for stage II and III, as well as some stage I, HER2-positive and triple-negative tumors. Neoadjuvant endocrine therapy (NET) strategies have also emerged from innovative trials for stage II and III estrogen receptor (ER)-positive/HER2-negative tumors, as in the ALTERNATE trial. From neoadjuvant trials, opportunities have emerged to de-escalate therapy on the basis of metrics of response to chemotherapy or hormonal therapy. Neoadjuvant therapy for early breast cancer is therefore emerging as a promising approach to accelerate new drug development, optimize treatment strategies, and (where appropriate) de-escalate neoadjuvant therapy.
Collapse
Affiliation(s)
| | - César A Herrera-Ponzanelli
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Diego Hinojosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Rafael Castillo
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Irwin Hernandez-Cruz
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
| | - Victor A Arrieta
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, México
- Department of Neurosurgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| | | | - Douglas Yee
- University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
187
|
Schmidt H, Zhaveri S, Valente C, Pisapati K, Pickholz E, Weltz S, Nayak A, Oza T, Corben A, Weltz C, Port E, Jaffer S. Response in breast vs axilla after neoadjuvant treatment and implications for nonoperative management of invasive breast cancer. Breast J 2021; 27:120-125. [PMID: 33393166 DOI: 10.1111/tbj.14125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/31/2022]
Abstract
Improved imaging and neoadjuvant chemotherapy (NAT) have led to higher pathologic complete response rates (pCR) in patients with invasive breast cancer. This has questioned the necessity of surgery and axillary lymph node (ALN) dissection in these patients. Prospective clinical trials are implementing extensive core biopsies of the tumor bed of patients with clinical complete response as a means to identify and spare them breast surgery. In addition, it is anticipated that patients with pCR are most likely going to have no or minimal disease in ALN as well. To verify the feasibility of these trials, we performed a pathologic analysis of all our patients who have undergone NAT from 2009 to present. Using pathology data base, we identified 362 patients treated with neoadjuvant chemotherapy followed by surgery. Clinical and pathologic information including gross and microscopic descriptions as well as biomarker status was collected. pCR was 50% for patients with negative ALN pretreatment but only 28% for patients with positive ALN at diagnosis. Despite achieving pCR in the breast, up to 10% of patients with positive ALN and 1% with negative ALN had persistent disease. Eight percent of patients that were presumed to have no ALN disease either clinically and or by imaging were found to have metastatic carcinoma in ALN. The metastases were predominantly (80%) <5 mm, and not palpable on physical examination and or due to biopsy sampling error. pCR in breast and ALN directly correlated with tumor size, ALN disease, and Her2 positive and triple negative receptor phenotype. In breast cancer patients who are node positive at time of diagnosis with pCR in the breast after neoadjuvant chemotherapy, residual lymph node disease was very uncommon. Further study is warranted to select patients who may avoid breast and axillary surgery post neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Hank Schmidt
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shruti Zhaveri
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Valente
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kereeti Pisapati
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eliana Pickholz
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Weltz
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anupma Nayak
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Twisha Oza
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adriana Corben
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina Weltz
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elisa Port
- Breast Surgical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shabnam Jaffer
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
188
|
Adjuvant and Neoadjuvant Treatment of Triple-Negative Breast Cancer With Chemotherapy. ACTA ACUST UNITED AC 2021; 27:41-49. [PMID: 33475292 DOI: 10.1097/ppo.0000000000000498] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
ABSTRACT Triple-negative breast cancer (TNBC) accounts for 15% to 20% of all invasive breast carcinomas and is defined by the lack of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Although TNBC is characterized by high rates of disease recurrence and worse survival, it is significantly more sensitive to chemotherapy as compared with other breast cancer subtypes. Accordingly, despite great efforts in the genomic characterization of TNBC, chemotherapy still represents the cornerstone of treatment. For the majority of patients with early-stage TNBC, sequential anthracycline- and taxane-based neoadjuvant chemotherapy (NACT) represents the standard therapeutic approach, with pathological complete response that strongly correlates with long-term survival outcomes. However, some issues about the optimal neoadjuvant regimen, as well as the effective role of chemotherapy in patients with residual disease after NACT, are still debated. Herein, we will review the current evidences that guide the use of (neo)adjuvant chemotherapy in patients with early-stage TNBC. Furthermore, we will discuss current controversies, including the incorporation of platinum compounds to the neoadjuvant backbone and the optimal treatment for patients with residual disease after NACT. Lastly, we will outline potential future directions that can guide treatment escalation and de-escalation, as well as the development of new therapies. In our view, the application of multi-omics technologies, liquid biopsy assays, and machine learning algorithms are strongly warranted to pave the way toward personalized anticancer treatment for early-stage TNBC.
Collapse
|
189
|
Cancer Detection and Quantification of Treatment Response Using Diffusion-Weighted MRI. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
190
|
Weng ZJ, Wu SX, Luo HS, Du ZS, Li XY, Lin JZ. Neoadjuvant Chemotherapy in Early Triple-Negative Breast Cancer: A Pairwise and Network Meta-Analysis of Pathological Complete Response. INQUIRY : A JOURNAL OF MEDICAL CARE ORGANIZATION, PROVISION AND FINANCING 2021; 58:469580211056213. [PMID: 34806458 PMCID: PMC8606982 DOI: 10.1177/00469580211056213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We performed a pairwise and network meta-analysis to compare pathological complete response (pCR) among neoadjuvant chemotherapy in patients with triple-negative breast cancer. We searched PubMed for randomized clinical trials between January 1, 2000 and December 1, 2020. Abstracts from meetings were also searched. A frequentist random-effect model was applied to compare pCR and toxicities. The P-score was used to rank treatment effects. Nineteen trials with 16 treatments and 7794 patients were included. On the basis of SoC, the addition of carboplatin (OR = 1.82, 95% CI, 1.24 to 2.68, P < .01) and the addition of checkpoint inhibitors (OR = 1.69, 95% CI, 1.23 to 2.32, P < .01) increased pCR in pairwise meta-analysis; compared with paclitaxel, nab-paclitaxel did not improve pCR rates (OR = 1.81, 95% CI, .80 to 4.12, P = .16). The anthracycline-sparing regimen led to similar pCR compared with the anthracycline-containing regimen (OR = 1.50, 95% CI, .82 to 2.76, P = .19). In network meta-analysis, the addition of carboplatin plus a PD-1 inhibitor (pembrolizumab), carboplatin plus bevacizumab, and carboplatin plus veliparib ranked as the top three treatments for achieving pCR, with corresponding P-scores of .91, .84, and .72, respectively. Among patients with homologous recombination deficiency, the addition of carboplatin (OR = 1.31, 95% CI, .69 to 2.50, P = .41) or carboplatin plus PARP inhibitors (OR = 1.19, 95% CI, .58 to 2.47, P = .63) did not increase pCR. For triple-negative breast cancer, combining carboplatin with taxane-anthracycline-containing neoadjuvant chemotherapy could be the standard of care, and the combination containing checkpoint inhibitor is promising. However, their role in long-term oncologic outcome remains to be determined.
Collapse
Affiliation(s)
- Zeng-Jie Weng
- Department of General Practice, Shantou Central Hospital, Shantou, China
| | - Sheng-Xi Wu
- Department of Radiation Oncology, Shantou Central Hospital, Shantou, China
| | - He-San Luo
- Department of Radiation Oncology, Shantou Central Hospital, Shantou, China
| | - Ze-Sen Du
- Department of Surgical Oncology, Shantou Central Hospital, Shantou, China
| | - Xu-Yuan Li
- Department of Medical Oncology, Shantou Central Hospital, Shantou, China
| | - Jia-Zhou Lin
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
191
|
Cao L, Zhang M, Yu X, Wang J, Li Y. Efficacy and safety of poly (ADP-ribose) polymerase inhibitors therapy for BRCA-mutated breast cancer: A systematic review and meta-analysis. J Cancer Res Ther 2021; 17:1672-1678. [DOI: 10.4103/jcrt.jcrt_2085_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
192
|
Asaoka M, Gandhi S, Ishikawa T, Takabe K. Neoadjuvant Chemotherapy for Breast Cancer: Past, Present, and Future. Breast Cancer (Auckl) 2020; 14:1178223420980377. [PMID: 33402827 PMCID: PMC7747102 DOI: 10.1177/1178223420980377] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Neoadjuvant chemotherapy (NAC) had been developed as a systematic approach before definitive surgery for the treatment of locally advanced or inoperable breast cancer such as inflammatory breast cancer in the past. In addition to its impact on surgery, the neoadjuvant setting has a benefit of providing the opportunity to monitor the individual drug response. Currently, the subject of NAC has expanded to include patients with early-stage, operable breast cancer because it is revealed that the achievement of a pathologic complete response (pCR) is associated with excellent long-term outcomes, especially in patients with aggressive phenotype breast cancer. In addition, this approach provides the unique opportunity to escalate adjuvant therapy in those with residual disease after NAC. Neoadjuvant chemotherapy in breast cancer is a rapidly evolving topic with tremendous interest in ongoing clinical trials. Here, we review the improvements and further challenges in the NAC setting in translational breast cancer research.
Collapse
Affiliation(s)
- Mariko Asaoka
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Shipra Gandhi
- Breast Medicine, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, Japan
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
193
|
Molecular Features and Clinical Management of Hereditary Gynecological Cancers. Int J Mol Sci 2020; 21:ijms21249504. [PMID: 33327492 PMCID: PMC7765001 DOI: 10.3390/ijms21249504] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
Hereditary gynecological cancers are caused by several inherited genes. Tumors that arise in the female reproductive system, such as ovaries and the uterus, overlap with hereditary cancers. Several hereditary cancer-related genes are important because they might lead to therapeutic targets. Treatment of hereditary cancers should be updated in line with the advent of various new methods of evaluation. Next-generation sequencing has led to rapid, economical genetic analyses that have prompted a concomitant and significant paradigm shift with respect to hereditary cancers. Molecular tumor profiling is an epochal method for determining therapeutic targets. Clinical treatment strategies are now being designed based on biomarkers based on tumor profiling. Furthermore, the National Comprehensive Cancer Network (NCCN) guidelines significantly changed the genetic testing process in 2020 to initially consider multi-gene panel (MGP) evaluation. Here, we reviewed the molecular features and clinical management of hereditary gynecological malignancies, such as hereditary breast and ovarian cancer (HBOC), and Lynch, Li–Fraumeni, Cowden, and Peutz–Jeghers syndromes. We also reviewed cancer-susceptible genes revealed by MGP tests.
Collapse
|
194
|
Ali RMM, McIntosh SA, Savage KI. Homologous recombination deficiency in breast cancer: Implications for risk, cancer development, and therapy. Genes Chromosomes Cancer 2020; 60:358-372. [DOI: 10.1002/gcc.22921] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/23/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Rayhaan M. M. Ali
- Patrick G Johnston Centre for Cancer Research Queen's University Belfast Belfast UK
| | - Stuart A. McIntosh
- Patrick G Johnston Centre for Cancer Research Queen's University Belfast Belfast UK
| | - Kienan I. Savage
- Patrick G Johnston Centre for Cancer Research Queen's University Belfast Belfast UK
| |
Collapse
|
195
|
Li W, Newitt DC, Gibbs J, Wilmes LJ, Jones EF, Arasu VA, Strand F, Onishi N, Nguyen AAT, Kornak J, Joe BN, Price ER, Ojeda-Fournier H, Eghtedari M, Zamora KW, Woodard SA, Umphrey H, Bernreuter W, Nelson M, Church AL, Bolan P, Kuritza T, Ward K, Morley K, Wolverton D, Fountain K, Lopez-Paniagua D, Hardesty L, Brandt K, McDonald ES, Rosen M, Kontos D, Abe H, Sheth D, Crane EP, Dillis C, Sheth P, Hovanessian-Larsen L, Bang DH, Porter B, Oh KY, Jafarian N, Tudorica A, Niell BL, Drukteinis J, Newell MS, Cohen MA, Giurescu M, Berman E, Lehman C, Partridge SC, Fitzpatrick KA, Borders MH, Yang WT, Dogan B, Goudreau S, Chenevert T, Yau C, DeMichele A, Berry D, Esserman LJ, Hylton NM. Predicting breast cancer response to neoadjuvant treatment using multi-feature MRI: results from the I-SPY 2 TRIAL. NPJ Breast Cancer 2020; 6:63. [PMID: 33298938 PMCID: PMC7695723 DOI: 10.1038/s41523-020-00203-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Dynamic contrast-enhanced (DCE) MRI provides both morphological and functional information regarding breast tumor response to neoadjuvant chemotherapy (NAC). The purpose of this retrospective study is to test if prediction models combining multiple MRI features outperform models with single features. Four features were quantitatively calculated in each MRI exam: functional tumor volume, longest diameter, sphericity, and contralateral background parenchymal enhancement. Logistic regression analysis was used to study the relationship between MRI variables and pathologic complete response (pCR). Predictive performance was estimated using the area under the receiver operating characteristic curve (AUC). The full cohort was stratified by hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) status (positive or negative). A total of 384 patients (median age: 49 y/o) were included. Results showed analysis with combined features achieved higher AUCs than analysis with any feature alone. AUCs estimated for the combined versus highest AUCs among single features were 0.81 (95% confidence interval [CI]: 0.76, 0.86) versus 0.79 (95% CI: 0.73, 0.85) in the full cohort, 0.83 (95% CI: 0.77, 0.92) versus 0.73 (95% CI: 0.61, 0.84) in HR-positive/HER2-negative, 0.88 (95% CI: 0.79, 0.97) versus 0.78 (95% CI: 0.63, 0.89) in HR-positive/HER2-positive, 0.83 (95% CI not available) versus 0.75 (95% CI: 0.46, 0.81) in HR-negative/HER2-positive, and 0.82 (95% CI: 0.74, 0.91) versus 0.75 (95% CI: 0.64, 0.83) in triple negatives. Multi-feature MRI analysis improved pCR prediction over analysis of any individual feature that we examined. Additionally, the improvements in prediction were more notable when analysis was conducted according to cancer subtype.
Collapse
Affiliation(s)
- Wen Li
- University of California, San Francisco, CA, USA
| | | | | | | | - Ella F Jones
- University of California, San Francisco, CA, USA
| | | | - Fredrik Strand
- University of California, San Francisco, CA, USA
- Karolinska Institute, Stockholm, Sweden
| | | | | | - John Kornak
- University of California, San Francisco, CA, USA
| | - Bonnie N Joe
- University of California, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Mark Rosen
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | - Pulin Sheth
- University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Karen Y Oh
- Oregon Health & Science University, Portland, OR, USA
| | - Neda Jafarian
- Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Wei T Yang
- University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Basak Dogan
- University of Texas, M.D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | - Don Berry
- Berry Consultants, LLC, Austin, TX, USA
| | | | | |
Collapse
|
196
|
Olaparib monotherapy as primary treatment in unselected triple negative breast cancer. Ann Oncol 2020; 32:240-249. [PMID: 33242536 DOI: 10.1016/j.annonc.2020.11.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/01/2020] [Accepted: 11/12/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The antitumor efficacy of PARP inhibitors (PARPi) for breast cancer patients harboring germline BRCA1/2 (gBRCA1/2) mutations is well established. While PARPi monotherapy was ineffective in patients with metastatic triple negative breast cancer (TNBC) wild type for BRCA1/2, we hypothesized that PARPi may be effective in primary TNBCs without previous chemotherapy exposure. PATIENTS AND METHODS In the phase II PETREMAC trial, patients with primary TNBC >2 cm received olaparib for up to 10 weeks before chemotherapy. Tumor biopsies collected before and after olaparib underwent targeted DNA sequencing (360 genes) and BRCA1 methylation analyses. In addition, BRCAness (multiplex ligation-dependent probe amplification), PAM50 gene expression, RAD51 foci, tumor-infiltrating lymphocytes (TILs) and PD-L1 analyses were performed on pretreatment samples. RESULTS The median pretreatment tumor diameter was 60 mm (range 25-112 mm). Eighteen out of 32 patients obtained an objective response (OR) to olaparib (56.3%). Somatic or germline mutations affecting homologous recombination (HR) were observed in 10/18 responders [OR 55.6%, 95% confidence interval (CI) 33.7-75.4] contrasting 1/14 non-responders (OR 7.1%; CI 1.3-31.5, P = 0.008). Among tumors without HR mutations, 6/8 responders versus 3/13 non-responders revealed BRCA1 hypermethylation (P = 0.03). Thus, 16/18 responders (88.9%, CI 67.2-96.9), in contrast to 4/14 non-responders (28.6%, CI 11.7-54.7, P = 0.0008), carried HR mutations and/or BRCA1 methylation. Excluding one gPALB2 and four gBRCA1/2 mutation carriers, 12/14 responders (85.7%, CI 60.1-96.0) versus 3/13 non-responders (23.1%, CI 8.2-50.3, P = 0.002) carried somatic HR mutations and/or BRCA1 methylation. In contrast to BRCAness signature or basal-like subtype, low RAD51 scores, high TIL or high PD-L1 expression all correlated to olaparib response. CONCLUSION Olaparib yielded a high clinical response rate in treatment-naïve TNBCs revealing HR deficiency, beyond germline HR mutations. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT02624973.
Collapse
|
197
|
Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol 2020; 32:229-239. [PMID: 33232761 PMCID: PMC9348585 DOI: 10.1016/j.annonc.2020.11.007] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/29/2020] [Accepted: 11/08/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) is strongly associated with favorable outcome. We examined the utility of serial circulating tumor DNA (ctDNA) testing for predicting pCR and risk of metastatic recurrence. PATIENTS AND METHODS Cell-free DNA (cfDNA) was isolated from 291 plasma samples of 84 high-risk early breast cancer patients treated in the neoadjuvant I-SPY 2 TRIAL with standard NAC alone or combined with MK-2206 (AKT inhibitor) treatment. Blood was collected at pretreatment (T0), 3 weeks after initiation of paclitaxel (T1), between paclitaxel and anthracycline regimens (T2), or prior to surgery (T3). A personalized ctDNA test was designed to detect up to 16 patient-specific mutations (from whole-exome sequencing of pretreatment tumor) in cfDNA by ultra-deep sequencing. The median follow-up time for survival analysis was 4.8 years. RESULTS At T0, 61 of 84 (73%) patients were ctDNA positive, which decreased over time (T1: 35%; T2: 14%; and T3: 9%). Patients who remained ctDNA positive at T1 were significantly more likely to have residual disease after NAC (83% non-pCR) compared with those who cleared ctDNA (52% non-pCR; odds ratio 4.33, P = 0.012). After NAC, all patients who achieved pCR were ctDNA negative (n = 17, 100%). For those who did not achieve pCR (n = 43), ctDNA-positive patients (14%) had a significantly increased risk of metastatic recurrence [hazard ratio (HR) 10.4; 95% confidence interval (CI) 2.3-46.6]; interestingly, patients who did not achieve pCR but were ctDNA negative (86%) had excellent outcome, similar to those who achieved pCR (HR 1.4; 95% CI 0.15-13.5). CONCLUSIONS Lack of ctDNA clearance was a significant predictor of poor response and metastatic recurrence, while clearance was associated with improved survival even in patients who did not achieve pCR. Personalized monitoring of ctDNA during NAC of high-risk early breast cancer may aid in real-time assessment of treatment response and help fine-tune pCR as a surrogate endpoint of survival.
Collapse
|
198
|
Ha R, Chin C, Karcich J, Liu MZ, Chang P, Mutasa S, Pascual Van Sant E, Wynn RT, Connolly E, Jambawalikar S. Prior to Initiation of Chemotherapy, Can We Predict Breast Tumor Response? Deep Learning Convolutional Neural Networks Approach Using a Breast MRI Tumor Dataset. J Digit Imaging 2020; 32:693-701. [PMID: 30361936 DOI: 10.1007/s10278-018-0144-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We hypothesize that convolutional neural networks (CNN) can be used to predict neoadjuvant chemotherapy (NAC) response using a breast MRI tumor dataset prior to initiation of chemotherapy. An institutional review board-approved retrospective review of our database from January 2009 to June 2016 identified 141 locally advanced breast cancer patients who (1) underwent breast MRI prior to the initiation of NAC, (2) successfully completed adriamycin/taxane-based NAC, and (3) underwent surgical resection with available final surgical pathology data. Patients were classified into three groups based on their NAC response confirmed on final surgical pathology: complete (group 1), partial (group 2), and no response/progression (group 3). A total of 3107 volumetric slices of 141 tumors were evaluated. Breast tumor was identified on first T1 postcontrast dynamic images and underwent 3D segmentation. CNN consisted of ten convolutional layers, four max-pooling layers, and dropout of 50% after a fully connected layer. Dropout, augmentation, and L2 regularization were implemented to prevent overfitting of data. Non-linear functions were modeled by a rectified linear unit (ReLU). Batch normalization was used between the convolutional and ReLU layers to limit drift of layer activations during training. A three-class neoadjuvant prediction model was evaluated (group 1, group 2, or group 3). The CNN achieved an overall accuracy of 88% in three-class prediction of neoadjuvant treatment response. Three-class prediction discriminating one group from the other two was analyzed. Group 1 had a specificity of 95.1% ± 3.1%, sensitivity of 73.9% ± 4.5%, and accuracy of 87.7% ± 0.6%. Group 2 (partial response) had a specificity of 91.6% ± 1.3%, sensitivity of 82.4% ± 2.7%, and accuracy of 87.7% ± 0.6%. Group 3 (no response/progression) had a specificity of 93.4% ± 2.9%, sensitivity of 76.8% ± 5.7%, and accuracy of 87.8% ± 0.6%. It is feasible for current deep CNN architectures to be trained to predict NAC treatment response using a breast MRI dataset obtained prior to initiation of chemotherapy. Larger dataset will likely improve our prediction model.
Collapse
Affiliation(s)
- Richard Ha
- Department of Radiology, Columbia University Irving Medical Center, 622 West 168th Street, PB-1-301, New York, NY, 10032, USA.
| | - Christine Chin
- Division of Radiation Oncology, Columbia University Medical Center, Presbyterian Hospital Building, 622 West 168th Street, Level B, New York, NY, 10032, USA
| | - Jenika Karcich
- Department of Radiology, Columbia University Irving Medical Center, 622 West 168th Street, PB-1-301, New York, NY, 10032, USA
| | - Michael Z Liu
- Department of Medical Physics, Columbia University Medical Center, 177 Ft. Washington Ave., Milstein Bldg Room 3-124B, New York, NY, 10032-3784, USA
| | - Peter Chang
- Department of Radiology, UC San Francisco Medical Center, 505 Parnassus Ave, San Francisco, CA, 94143, USA
| | - Simukayi Mutasa
- Department of Radiology, Columbia University Irving Medical Center, 622 West 168th Street, PB-1-301, New York, NY, 10032, USA
| | - Eduardo Pascual Van Sant
- Department of Radiology, Columbia University Irving Medical Center, 622 West 168th Street, PB-1-301, New York, NY, 10032, USA
| | - Ralph T Wynn
- Department of Radiology, Columbia University Irving Medical Center, 622 West 168th Street, PB-1-301, New York, NY, 10032, USA
| | - Eileen Connolly
- Division of Radiation Oncology, Columbia University Medical Center, Presbyterian Hospital Building, 622 West 168th Street, Level B, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Medical Physics, Columbia University Medical Center, 177 Ft. Washington Ave., Milstein Bldg Room 3-124B, New York, NY, 10032-3784, USA
| |
Collapse
|
199
|
Sun W, Li J, Zhang Z, Su X. Gastrointestinal events with PARP inhibitors in cancer patients: A meta-analysis of phase II/III randomized controlled trials. J Clin Pharm Ther 2020; 46:241-255. [PMID: 33135237 DOI: 10.1111/jcpt.13300] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 11/28/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE PARP inhibitors are currently one of the most promising PARP targeted drugs for patients with certain types of cancer. Gastrointestinal (GI) events are common adverse events for all PARP inhibitors. We conducted this meta-analysis of randomized controlled trials (RCTs) to fully investigate the incidence and the relative risk of GI events in cancer patients receiving PARP inhibitors. STUDY DESIGN Randomized controlled trials in cancer patients treated with PARP inhibitors were retrieved, and the systematic evaluation was conducted. Embase and PubMed/Medline were searched for articles published till July 2020. RESULTS Twenty-nine RCTs and 9529 patients were included. The present meta-analysis suggests that the use of PARP inhibitors significantly increases the risk of developing all-grade nausea (RR, 1.46; 95% CI, 1.29-1.66; p < .00001), vomiting (RR, 1.39; 95% CI, 1.17-1.64; p = .0001), diarrhoea (RR, 1.14; 95% CI, 1.06-1.23; p = .0003) and decreased appetite (RR, 1.24; 95% CI, 1.14-1.36; p < .00001), but not for constipation. And the use of these agents significantly increased the risk of high-grade nausea (RR, 1.99; 95% CI, 1.44-2.74; p < .0001), vomiting (RR, 1.54; 95% CI, 1.11-2.14; p = .01) and decreased appetite (RR, 2.03; 95% CI, 1.22-3.40; p = .007), except for diarrhoea and constipation. Nausea was the most common GI event for these agents. Patients receiving veliparib were associated with a relatively lower risk of all-grade nausea and vomiting. Patients with ovarian cancer tend to have a higher risk of all-grade nausea and vomiting than those with non-ovarian cancer. The risk of all-grade nausea and vomiting tended to be higher when PARP inhibitors treatment was longer. WHAT IS NEW AND CONCLUSION PARP inhibitors were associated with a significant increased risk of GI events. Clinicians should be aware of these risks and perform regular monitoring.
Collapse
Affiliation(s)
- Wenxia Sun
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Chengdu University, Chengdu, China
| | - Jing Li
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Zhifeng Zhang
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| | - Xueyan Su
- College of Pharmacy, Southwest Minzu University, Chengdu, China
| |
Collapse
|
200
|
Hyland CJ, Varghese F, Yau C, Beckwith H, Khoury K, Varnado W, Hirst GL, Flavell RR, Chien AJ, Yee D, Isaacs CJ, Forero-Torres A, Esserman LJ, Melisko ME. Use of 18F-FDG PET/CT as an Initial Staging Procedure for Stage II-III Breast Cancer: A Multicenter Value Analysis. J Natl Compr Canc Netw 2020; 18:1510-1517. [PMID: 33152704 DOI: 10.6004/jnccn.2020.7598] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 05/25/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Metastatic staging imaging is not recommended for asymptomatic patients with stage I-II breast cancer. Greater distant metastatic disease risk may warrant baseline imaging in patients with stage II-III with high-risk biologic subtypes. NCCN Guidelines recommend considering CT of the chest, abdomen, and pelvis (CT CAP) and bone scan in appropriate patients. CT CAP and bone scan are considered standard of care (SoC), although PET/CT is a patient-centered alternative. METHODS Data were available for 799 high-risk patients with clinical stage II-III disease who initiated screening for the I-SPY2 trial at 4 institutions. A total of 564 complete records were reviewed to compare PET/CT versus SoC. Costs were determined from the payer perspective using the national 2018 Medicare Physician Fee Schedule and representative reimbursements to the University of California, San Francisco (UCSF). Incremental cost-effectiveness ratio (ICER) measured cost of using PET/CT per percent of patients who avoided a false-positive (FP). RESULTS The de novo metastatic disease rate was 4.6%. Imaging varied across the 4 institutions (P<.0001). The FP rate was higher using SoC versus PET/CT (22.1% vs 11.1%; P=.0009). Mean time between incidental finding on baseline imaging to FP determination was 10.8 days. Mean time from diagnosis to chemotherapy initiation was 44.3 days with SoC versus 37.5 days with PET/CT (P=.0001). Mean cost per patient was $1,132 (SoC) versus $1,477 (PET/CT) using the Medicare Physician Fee Schedule, with an ICER of $31. Using representative reimbursements to UCSF, mean cost per patient was $1,236 (SoC) versus $1,073 (PET/CT) for Medicare, and $3,083 (SoC) versus $1,656 (PET/CT) for a private payer, with ICERs of -$15 and -$130, respectively. CONCLUSIONS Considerable variation exists in metastatic staging practices. PET/CT reduced FP risk by half and decreased workup of incidental findings, allowing for earlier treatment start. PET/CT may be cost-effective, and at one institution was shown to be cost-saving. Better alignment is needed between hospital pricing strategies and payer coverage policies to deliver high-value care.
Collapse
Affiliation(s)
- Colby J Hyland
- 1University of California, San Francisco, San Francisco, California
| | - Flora Varghese
- 1University of California, San Francisco, San Francisco, California
| | - Christina Yau
- 1University of California, San Francisco, San Francisco, California
| | | | | | - William Varnado
- 4University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Gillian L Hirst
- 1University of California, San Francisco, San Francisco, California
| | - Robert R Flavell
- 5Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - A Jo Chien
- 1University of California, San Francisco, San Francisco, California
| | - Douglas Yee
- 2University of Minnesota, Minneapolis, Minnesota
| | | | | | - Laura J Esserman
- 1University of California, San Francisco, San Francisco, California
| | | |
Collapse
|