Searched for: in-biosketch:true
person:adamss04
Prognostic Value of Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancers From Two Phase III Randomized Adjuvant Breast Cancer Trials: ECOG 2197 and ECOG 1199
Adams, Sylvia; Gray, Robert J; Demaria, Sandra; Goldstein, Lori; Perez, Edith A; Shulman, Lawrence N; Martino, Silvana; Wang, Molin; Jones, Vicky E; Saphner, Thomas J; Wolff, Antonio C; Wood, William C; Davidson, Nancy E; Sledge, George W; Sparano, Joseph A; Badve, Sunil S
PURPOSE: Recent studies suggest that tumor-infiltrating lymphocytes (TILs) are associated with disease-free (DFS) and overall survival (OS) in operable triple-negative breast cancer (TNBC). We seek to validate the prognostic impact of TILs in primary TNBCs in two adjuvant phase III trials conducted by the Eastern Cooperative Oncology Group (ECOG). PATIENTS AND METHODS: Full-face hematoxylin and eosin-stained sections of 506 tumors from ECOG trials E2197 and E1199 were evaluated for density of TILs in intraepithelial (iTILs) and stromal compartments (sTILs). Patient cases of TNBC from E2197 and E1199 were randomly selected based on availability of sections. For the primary end point of DFS, association with TIL scores was determined by fitting proportional hazards models stratified on study. Secondary end points were OS and distant recurrence-free interval (DRFI). Reporting recommendations for tumor marker prognostic studies criteria were followed, and all analyses were prespecified. RESULTS: The majority of 481 evaluable cancers had TILs (sTILs, 80%; iTILs, 15%). With a median follow-up of 10.6 years, higher sTIL scores were associated with better prognosis; for every 10% increase in sTILs, a 14% reduction of risk of recurrence or death (P = .02), 18% reduction of risk of distant recurrence (P = .04), and 19% reduction of risk of death (P =.01) were observed. Multivariable analysis confirmed sTILs to be an independent prognostic marker of DFS, DRFI, and OS. CONCLUSION: In two national randomized clinical trials using contemporary adjuvant chemotherapy, we confirm that stromal lymphocytic infiltration constitutes a robust prognostic factor in TNBCs. Studies assessing outcomes and therapeutic efficacies should consider stratification for this parameter.
PMCID:4162494
PMID: 25071121
ISSN: 0732-183x
CID: 1089992
MAGE-specific T cells detected directly ex-vivo correlate with complete remission in metastatic breast cancer patients after sequential immune-endocrine therapy
Janosky, Maxwell; Sabado, Rachel L; Cruz, Crystal; Vengco, Isabelita; Hasan, Farah; Winer, Arthur; Moy, Linda; Adams, Sylvia
Studies suggest that conventional cancer therapies given after immunotherapy (IT) can boost antitumor immunity and possibly improve response rates and progression-free survival. We report two cases of metastatic breast cancer with durable complete responses (CRs) after sequential IT and endocrine therapy. Immune analyses of these long-term disease-free breast cancer patients previously treated with imiquimod (IMQ) suggest in-situ vaccination is achieved by topical application of the TLR-7 agonist directly onto tumors. Furthermore, IT-induced antigen-specific T cells were expanded by subsequent endocrine therapy and correlated with response, persisting > 2 years. Our findings therefore suggest that the induction/boosting of polyfunctional tumor antigen-specific T in response to sequential immune endocrine therapy and detected directly ex-vivo can serve as a peripheral blood biomarker for true clinical benefit.
PMCID:5569937
PMID: 28837000
ISSN: 2051-1426
CID: 2676082
Phase 2 trial of everolimus and carboplatin combination in patients with triple negative metastatic breast cancer
Singh, Jasmeet; Novik, Yelena; Stein, Stacey; Volm, Matthew; Meyers, Marlene; Smith, Julia; Omene, Coral; Speyer, James; Schneider, Robert; Jhaveri, Komal; Formenti, Silvia; Kyriakou, Victoria; Joseph, Benson; Goldberg, Judith D; Li, Xiaochun; Adams, Sylvia; Tiersten, Amy
INTRODUCTION: Rapamycin acts synergistically with platinum agents to induce apoptosis and inhibit proliferation in breast cancer cell lines. Combination of everolimus also known as RAD001 (oral mammalian target of rapamycin (mTOR) inhibitor) and carboplatin may have activity in metastatic triple-negative breast cancer (TNBC). METHODS: The primary objective of this study was to determine clinical benefit rate (CBR), that is (complete remission (CR) + partial remission (PR) + stable disease (SD) lasting >/=6 months) and the toxicity of everolimus/carboplatin in women with metastatic TNBC. Prior carboplatin was allowed. Treatment consisted of intravenous carboplatin area under the curve (AUC) 6 (later decreased to AUC 5 and subsequently to AUC 4) every 3 weeks with daily 5 mg everolimus. RESULTS: We enrolled 25 patients in this study. Median age was 58 years. There were one CR, six PRs, seven SDs and eight PDs (progression of disease). CBR was 36% (95% confidence interval (CI) 21.1 to 57.4%). One SD was achieved in a patient progressing on single agent carboplatin. The median progression free survival (PFS) was 3 months (95% CI 1.6 to 4.6 months) and overall survival (OS) was 16.6 months (95% CI 7.3 months to not reached). There were seven patients (28%) with >/= grade 3 thrombocytopenia; three (12%) with grade 3 neutropenia (no bleeding/febrile neutropenia) and one (4%) with grade 3 anemia. Greater hematological toxicity was seen in the first seven patients treated with carboplatin AUC5/6. After the amendment for starting dose of carboplatin to AUC 4, the regimen was well tolerated with only one out of 18 patients with grade 3 neutropenia and two patients with grade 3 thrombocytopenia. There was only one case of mucositis. CONCLUSION: Everolimus-carboplatin was efficacious in metastatic TNBC. Dose limiting hematological toxicity was observed when AUC5/6 of carboplatin was combined with everolimus. However, carboplatin AUC 4 was well tolerated in combination with everolimus with continuing responses. TRIAL REGISTRATIONS: ClinicalTrials.gov NCT01127763.
PMCID:4053575
PMID: 24684785
ISSN: 1465-542x
CID: 1583872
Clinical Cancer Advances 2013: Annual Report on Progress Against Cancer from the American Society of Clinical Oncology
Patel, Jyoti D; Krilov, Lada; Adams, Sylvia; Aghajanian, Carol; Basch, Ethan; Brose, Marcia S; Carroll, William L; de Lima, Marcos; Gilbert, Mark R; Kris, Mark G; Marshall, John L; Masters, Gregory A; O'Day, Steven J; Polite, Blase; Schwartz, Gary K; Sharma, Sunil; Thompson, Ian; Vogelzang, Nicholas J; Roth, Bruce J
A MESSAGE FROM ASCO'S PRESIDENT: Since its founding in 1964, the American Society of Clinical Oncology (ASCO) has been committed to improving cancer outcomes through research and the delivery of quality care. Research is the bedrock of discovering better treatments--providing hope to the millions of individuals who face a cancer diagnosis each year. The studies featured in "Clinical Cancer Advances 2013: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology" represent the invaluable contributions of thousands of patients who participate in clinical trials and the scientists who conduct basic and clinical research. The insights described in this report, such as how cancers hide from the immune system and why cancers may become resistant to targeted drugs, enable us to envision a future in which cancer will be even more controllable and preventable. The scientific process is thoughtful, deliberate, and sometimes slow, but each advance, while helping patients, now also points toward new research questions and unexplored opportunities. Both dramatic and subtle breakthroughs occur so that progress against cancer typically builds over many years. Success requires vision, persistence, and a long-term commitment to supporting cancer research and training. Our nation's longstanding investment in federally funded cancer research has contributed significantly to a growing array of effective new treatments and a much deeper understanding of the drivers of cancer. But despite this progress, our position as a world leader in advancing medical knowledge and our ability to attract the most promising and talented investigators are now threatened by an acute problem: Federal funding for cancer research has steadily eroded over the past decade, and only 15% of the ever-shrinking budget is actually spent on clinical trials. This dismal reality threatens the pace of progress against cancer and undermines our ability to address the continuing needs of our patients. Despite this extremely challenging economic environment, we continue to make progress. Maintaining and accelerating that progress require that we keep our eyes on the future and pursue a path that builds on the stunning successes of the past. We must continue to show our policymakers the successes in cancer survival and quality of life (QOL) they have enabled, emphasizing the need to sustain our national investment in the remarkably productive US cancer research enterprise. We must also look to innovative methods for transforming how we care for-and learn from-patients with cancer. Consider, for example, that fewer than 5% of adult patients with cancer currently participate in clinical trials. What if we were able to draw lessons from the other 95%? This possibility led ASCO this year to launch CancerLinQ, a groundbreaking health information technology initiative that will provide physicians with access to vast quantities of clinical data about real-world patients and help achieve higher quality, higher value cancer care. As you read the following pages, I hope our collective progress against cancer over the past year inspires you. More importantly, I hope the pride you feel motivates you to help us accelerate the pace of scientific advancement. Clifford A. Hudis, MD, FACP President American Society of Clinical Oncology.
PMID: 24327669
ISSN: 0732-183x
CID: 811012
Distinct breast cancer subtypes in women with early-onset disease across races
Singh, Mandeep; Ding, Yi; Zhang, Li-Ying; Song, Dong; Gong, Yun; Adams, Sylvia; Ross, Dara S; Wang, Jin-Hua; Grover, Shruti; Doval, Dinesh Chandra; Shao, Charles; He, Zi-Li; Chang, Victor; Chin, Warren W; Deng, Fang-Ming; Singh, Baljit; Zhang, David; Xu, Ru-Liang; Lee, Peng
BACKGROUND: Racial disparities among breast cancer (BCa) patients are known but not well studied in early-onset BCa. We analyzed molecular subtypes in early-onset BCa across five major races. METHODS: A total of 2120 cases were included from non-Hispanic White (NHW), African American (AA) and Hispanic, Chinese and Indian. Based on ER, PR and HER-2 status, BCa was classified into 4 intrinsic subtypes as Luminal A, Luminal B, HER2/neu overexpression and Triple negative BCa (TNBC) subtypes. Data was stratified according to race and age as younger/early-onset group (40-years and younger) and older group (50-years and older). RESULTS: In early-onset BCa, incidence of TNBC was significantly higher (p = 0.0369) in Indian women followed by AA, Hispanic, NHW and Chinese women. Incidence of Her2 over-expression subtype also was highest in Indian women, followed by Hispanic, Chinese, AA and NHW women. In contrast, Luminal B subtype was most significantly higher in AA women (p = 0.0000) followed by NHW (p = 0.0002), Chinese (p = 0.0003), Hispanic (0.0128) and Indian (p = 0.0468) women. Luminal A subtype was most significantly reduced in Indian women (p = 0.0113) followed by Hispanic, AA, NHW and Chinese women. These results were based on statistical analysis with the mean of older group populations. CONCLUSIONS: These results show significant disparities in receptor subtypes across races. This study will contribute in developing optimal clinical trial protocols and personalized management strategies for early-onset BCa patients.
PMCID:4106652
PMID: 25057437
ISSN: 2156-6976
CID: 1268482
The TLR7 agonist imiquimod as an adjuvant for radiotherapy-elicited in situ vaccination against breast cancer
Demaria, Sandra; Vanpouille-Box, Claire; Formenti, Silvia C; Adams, Sylvia
Radiotherapy can convert malignant cells into an in situ anticancer vaccine, but is often inadequate at generating sufficient pro-inflammatory signals to optimally activate innate and adaptive immune responses. Topical imiquimod is a powerful pro-inflammatory agent with clinical activity against superficial skin cancers. These two modalities appear to complement each other, hence achieving local and systemic tumor control.
PMCID:3881106
PMID: 24404422
ISSN: 2162-4011
CID: 763592
Clinical cancer advances 2012: annual report on progress against cancer from the american society of clinical oncology
Roth, Bruce J; Krilov, Lada; Adams, Sylvia; Aghajanian, Carol A; Bach, Peter; Braiteh, Fadi; Brose, Marcia S; Ellis, Lee M; Erba, Harry; George, Daniel J; Gilbert, Mark R; Jacobson, Joseph O; Larsen, Eric C; Lichtman, Stuart M; Partridge, Ann H; Patel, Jyoti D; Quinn, David I; Robison, Leslie L; von Roenn, Jamie H; Samlowski, Wolfram; Schwartz, Gary K; Vogelzang, Nicholas J
A MESSAGE FROM ASCO'S PRESIDENT I am delighted to present you with "Clinical Cancer Advances 2012: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology." The American Society of Clinical Oncology (ASCO) uses this opportunity each year to share the steady progress occurring in our understanding and treatment of cancer. For 2012, we offer again an inspiring perspective on clinical cancer advances over the past year, but with a cautionary note: if current threats to federal funding materialize, future progress in cancer research will be seriously undermined. Continued progress against cancer. As you read the following pages of this report, I hope you will share my unabashed enthusiasm-and pride-in how far we have come. To appreciate what this progress has meant to the millions of people who receive a cancer diagnosis each year, consider the following: (1) two of three people in the United States live at least 5 years after a cancer diagnosis (up from roughly one of two in the 1970s); (2) the nation's cancer death rate has dropped 18% since the early 1990s, reversing decades of increases; and (3) individuals with cancer are increasingly able to live active, fulfilling lives because of better management of symptoms and treatments with fewer adverse effects. Importance of clinical cancer trials. These dramatic trends-and the advances highlighted in this report-would have been unthinkable without the engine that drives life-saving cancer treatment: clinical cancer research. Advances in technology and in our knowledge of how patient-specific molecular characteristics of the tumor and its environment fuel the growth of cancer have brought new hope to patients. Clinical trials are the key to translating cutting-edge laboratory discoveries into treatments that extend and improve the lives of those with cancer. But progress is only part of the story. Cancer remains a challenge, with many cancers undetected until their latest stages and others resisting most attempts at treatment. Tragically, cancer still kills more than 500,000 people in the United States every year, and its global burden is growing rapidly. Bridges to better care. To conquer cancer, we need to build bridges to the future-bridges that will get scientific advances to the patient's bedside quicker, bridges that will enable us to share information and learn what works in real time, and bridges that will improve care for all patients around the world. At ASCO, we recognize the unique role that oncologists must play. ASCO's "Accelerating Progress Against Cancer: Blueprint for Transforming Clinical and Translational Cancer Research,"(1) published last year, presents our vision and recommendations to make cancer research and patient care vastly more targeted, more efficient, and more effective. We have also launched a groundbreaking initiative, CancerLinQ, that aims to improve cancer care and speed research by drawing insights from the vast pool of data on patients in real-world settings. Renewing a national commitment to cancer research. We are on the threshold of major advances in cancer prevention, detection, and treatment-but only if, as a nation, we remain committed to this critical endeavor. The federally funded cancer research system is currently under threat by larger federal budget concerns. Clearly, Congress faces a complex budget environment, but now is not the time to retreat from our nation's commitment to conquering a disease that affects nearly all of us. Bold action must be taken to ensure that we can take full advantage of today's scientific and technologic opportunities. Please join me in celebrating our nation's progress against cancer and in recommitting ourselves to supporting cancer research. Millions of lives depend on it. Sandra M. Swain, MD President American Society of Clinical Oncology.
PMID: 23213095
ISSN: 0732-183x
CID: 214312
Long Chain Fatty Acyl-CoA Synthetase 4 Is a Biomarker for and Mediator of Hormone Resistance in Human Breast Cancer
Wu, Xinyu; Li, Yirong; Wang, Jinhua; Wen, Xin; Marcus, Max T; Daniels, Garrett; Zhang, David Y; Ye, Fei; Wang, Ling Hang; Du, Xinxin; Adams, Sylvia; Singh, Baljit; Zavadil, Jiri; Lee, Peng; Monaco, Marie E
The purpose of this study was to determine the role of long-chain fatty acyl-CoA synthetase 4 (ACSL4) in breast cancer. Public databases were utilized to analyze the relationship between ACSL4 mRNA expression and the presence of steroid hormone and human epidermal growth factor receptor 2 (HER2) in both breast cancer cell lines and tissue samples. In addition, cell lines were utilized to assess the consequences of either increased or decreased levels of ACSL4 expression. Proliferation, migration, anchorage-independent growth and apoptosis were used as biological end points. Effects on mRNA expression and signal transduction pathways were also monitored. A meta-analysis of public gene expression databases indicated that ACSL4 expression is positively correlated with a unique subtype of triple negative breast cancer (TNBC), characterized by the absence of androgen receptor (AR) and therefore referred to as quadruple negative breast cancer (QNBC). Results of experiments in breast cancer cell lines suggest that simultaneous expression of ACSL4 and a receptor is associated with hormone resistance. Forced expression of ACSL4 in ACSL4-negative, estrogen receptor alpha (ER)-positive MCF-7 cells resulted in increased growth, invasion and anchorage independent growth, as well as a loss of dependence on estrogen that was accompanied by a reduction in the levels of steroid hormone receptors. Sensitivity to tamoxifen, triacsin C and etoposide was also attenuated. Similarly, when HER2-positive, ACSL4-negative, SKBr3 breast cancer cells were induced to express ACSL4, the proliferation rate increased and the apoptotic effect of lapatinib was reduced. The growth stimulatory effect of ACSL4 expression was also observed in vivo in nude mice when MCF-7 control and ACSL4-expressing cells were utilized to induce tumors. Our data strongly suggest that ACSL4 can serve as both a biomarker for, and mediator of, an aggressive breast cancer phenotype.
PMCID:3796543
PMID: 24155918
ISSN: 1932-6203
CID: 598522
Topical TLR7 agonist imiquimod can induce immune-mediated rejection of skin metastases in patients with breast cancer
Adams, Sylvia; Kozhaya, Lina; Martiniuk, Frank; Meng, Tze-Chiang; Chiriboga, Luis; Liebes, Leonard; Hochman, Tsivia; Shuman, Nicholas; Axelrod, Deborah; Speyer, James; Novik, Yelena; Tiersten, Amy; Goldberg, Judith D; Formenti, Silvia C; Bhardwaj, Nina; Unutmaz, Derya; Demaria, Sandra
PURPOSE: Skin metastases of breast cancer remain a therapeutic challenge. Toll-like receptor 7 agonist imiquimod is an immune response modifier and can induce immune-mediated rejection of primary skin malignancies when topically applied. Here we tested the hypothesis that topical imiquimod stimulates local antitumor immunity and induces the regression of breast cancer skin metastases. EXPERIMENTAL DESIGN: A prospective clinical trial was designed to evaluate the local tumor response rate of breast cancer skin metastases treated with topical imiquimod, applied 5 d/wk for 8 weeks. Safety and immunologic correlates were secondary objectives. RESULTS: Ten patients were enrolled and completed the study. Imiquimod treatment was well tolerated, with only grade 1 to 2 transient local and systemic side effects consistent with imiquimod's immunomodulatory effects. Two patients achieved a partial response [20%; 95% confidence interval (CI), 3%-56%]. Responders showed histologic tumor regression with evidence of an immune-mediated response, showed by changes in the tumor lymphocytic infiltrate and locally produced cytokines. CONCLUSION: Topical imiquimod is a beneficial treatment modality for breast cancer metastatic to skin/chest wall and is well tolerated. Importantly, imiquimod can promote a proimmunogenic tumor microenvironment in breast cancer. Preclinical data generated by our group suggest superior results with a combination of imiquimod and ionizing radiation and we are currently testing in patients whether the combination can further improve antitumor immune and clinical responses.
PMCID:3580198
PMID: 22767669
ISSN: 1078-0432
CID: 1395722
Synergy of Topical Toll-like Receptor 7 Agonist with Radiation and Low-Dose Cyclophosphamide in a Mouse Model of Cutaneous Breast Cancer
Dewan, M Zahidunnabi; Vanpouille-Box, Claire; Kawashima, Noriko; Dinapoli, Sara; Babb, James S; Formenti, Silvia C; Adams, Sylvia; Demaria, Sandra
PURPOSE: This study tested the hypothesis that topical Toll-like receptor (TLR) 7 agonist imiquimod promotes antitumor immunity and synergizes with other treatments in a model of skin-involving breast cancer. EXPERIMENTAL DESIGN: TSA mouse breast carcinoma cells were injected s.c. into syngeneic mice. Imiquimod 5% or placebo cream was applied topically on the shaved skin overlying tumors three times/wk. In some experiments, local ionizing radiation therapy (RT) was delivered to the tumor in three fractions of 8 Gy, given on consecutive days. Cyclophosphamide was given intraperitoneally (i.p.) in one dose of 2 mg/mouse. Mice were followed for tumor growth and survival. RESULTS: Treatment with imiquimod significantly inhibited tumor growth, an effect that was associated with increased tumor infiltration by CD11c(+), CD4(+), and CD8(+) cells, and abolished by depletion of CD8(+) cells. Administration of imiquimod in combination with RT enhanced significantly tumor response compared with either treatment alone (P < 0.005), and 11% to 66% of irradiated tumors completely regressed. Importantly, the addition of topical imiquimod also resulted in growth inhibition of a secondary tumor outside of the radiation field. Low-dose cyclophosphamide given before start of treatment with imiquimod and RT further improved tumor inhibition and reduced tumor recurrence. Mice that remained tumor-free rejected a tumorigenic inoculum of TSA cells, showing long-term immunologic memory. CONCLUSIONS: Topical imiquimod inhibits tumor growth and synergizes with RT. Addition of cyclophosphamide further increases the therapeutic effect and induces protective immunologic memory, suggesting that this combination is a promising strategy for cutaneous breast cancer metastases. Clin Cancer Res; 18(24); 6668-78. (c)2012 AACR.
PMCID:3525760
PMID: 23048078
ISSN: 1078-0432
CID: 203832