58 results on '"Lyndsay N. Harris"'
Search Results
2. Reduction of Global H3K27me3 Enhances HER2/ErbB2 Targeted Therapy
- Author
-
Alison Hirukawa, Salendra Singh, Jarey Wang, Jonathan P. Rennhack, Matthew Swiatnicki, Virginie Sanguin-Gendreau, Dongmei Zuo, Kamilia Daldoul, Cynthia Lavoie, Morag Park, Eran R. Andrechek, Thomas F. Westbrook, Lyndsay N. Harris, Vinay Varadan, Harvey W. Smith, and William J. Muller
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Monoclonal antibodies (mAbs) targeting the oncogenic receptor tyrosine kinase ERBB2/HER2, such as Trastuzumab, are the standard of care therapy for breast cancers driven by ERBB2 overexpression and activation. However, a substantial proportion of patients exhibit de novo resistance. Here, by comparing matched Trastuzumab-naive and post-treatment patient samples from a neoadjuvant trial, we link resistance with elevation of H3K27me3, a repressive histone modification catalyzed by polycomb repressor complex 2 (PRC2). In ErbB2+ breast cancer models, PRC2 silences endogenous retroviruses (ERVs) to suppress anti-tumor type-I interferon (IFN) responses. In patients, elevated H3K27me3 in tumor cells following Trastuzumab treatment correlates with suppression of interferon-driven viral defense gene expression signatures and poor response. Using an immunocompetent model, we provide evidence that EZH2 inhibitors promote interferon-driven immune responses that enhance the efficacy of anti-ErbB2 mAbs, suggesting the potential clinical benefit of epigenomic reprogramming by H3K27me3 depletion in Trastuzumab-resistant disease. : Hirukawa et al. link Trastuzumab resistance in ErbB2+ breast cancers with activity of the methyltransferase EZH2, a key epigenetic regulator. By silencing retrotransposons, EZH2 suppresses type-I interferon signaling to limit immune surveillance. Retrotransposon de-repression following EZH2 inhibition triggers interferon responses and sensitizes immunocompetent in vivo models to ErbB2 antibody therapy. Keywords: ErbB2/HER2, breast cancer, Polycomb Repressor Complex 2, PRC2, epigenetics, Trastuzumab resistance, endogenous retroviruses, immune surveillance, type I interferon signaling, transcriptional silencing
- Published
- 2019
- Full Text
- View/download PDF
3. 'A rising tide lifts all boats': establishing a multidisciplinary genomic tumor board for breast cancer patients with advanced disease
- Author
-
Michelle L. McGowan, Roselle S. Ponsaran, Paula Silverman, Lyndsay N. Harris, and Patricia A. Marshall
- Subjects
Breast cancer ,Genomics ,Qualitative research ,Patient care ,Ethics ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Research suggests that multidisciplinary genomic tumor boards (MGTB) can inform cancer patient care, though little is known about factors influencing how MGTBs interpret genomic test results, make recommendations, and perceive the utility of this approach. This study’s objective was to observe, describe, and assess the establishment of the Breast Multidisciplinary Genomic Tumor Board, the first MGTB focused on interpreting genomic test results for breast cancer patients with advanced disease. Methods We conducted a qualitative case study involving participant observation at monthly MGTB meetings from October 2013 through November 2014 and interviews with 12 MGTB members. We analyzed social dynamics and interactions within the MGTB regarding interpretation of genomic findings and participants’ views on effectiveness of the MGTB in using genomics to inform patient care. Results Twenty-two physicians, physician-scientists, basic scientists, bioethicists, and allied care professionals comprised the MGTB. The MGTB reviewed FoundationOne™ results for 40 metastatic breast cancer patients. Based on findings, the board mostly recommended referring patients to clinical trials (34) and medical genetics (15), and Food and Drug Administration-approved (FDA) breast cancer therapies (13). Though multidisciplinary, recommendations were driven by medical oncologists. Interviewees described providing more precise care recommendations and professional development as advantages and the limited actionability of genomic test results as a challenge for the MGTB. Conclusions Findings suggest both feasibility and desirability of pooling professional expertise in genomically-guided breast cancer care and challenges to institutionalizing a Breast MGTB, specifically in promoting interdisciplinary contributions and managing limited actionability of genomic test results for patients with advanced disease.
- Published
- 2016
- Full Text
- View/download PDF
4. Efficacy and Safety of Trastuzumab as a Single Agent in First-Line Treatment of HER2-Overexpressing Metastatic Breast Cancer
- Author
-
Charles L, Vogel, Melody A, Cobleigh, Debu, Tripathy, John C, Gutheil, Lyndsay N, Harris, Louis, Fehrenbacher, Dennis J, Slamon, Maureen, Murphy, William F, Novotny, Michael, Burchmore, Steven, Shak, Stanford J, Stewart, and Michael, Press
- Subjects
Adult ,Aged, 80 and over ,Cancer Research ,Gene Amplification ,Antibodies, Monoclonal ,Antineoplastic Agents ,Breast Neoplasms ,Genes, erbB-2 ,Middle Aged ,Trastuzumab ,Antibodies, Monoclonal, Humanized ,Immunohistochemistry ,Treatment Outcome ,Oncology ,Quality of Life ,Humans ,Female ,Safety ,skin and connective tissue diseases ,In Situ Hybridization ,Aged - Abstract
PURPOSE To evaluate the efficacy and safety of first-line, single-agent trastuzumab in women with HER2-overexpressing metastatic breast cancer. PATIENTS AND METHODS One hundred fourteen women with HER2-overexpressing metastatic breast cancer were randomized to receive first-line treatment with trastuzumab 4 mg/kg loading dose, followed by 2 mg/kg weekly, or a higher 8 mg/kg loading dose, followed by 4 mg/kg weekly. RESULTS The objective response rate was 26% (95% confidence interval [CI], 18.2% to 34.4%), with seven complete and 23 partial responses. Response rates in 111 assessable patients with 3+ and 2+ HER2 overexpression by immunohistochemistry (IHC) were 35% (95% CI, 24.4% to 44.7%) and none (95% CI, 0% to 15.5%), respectively. The clinical benefit rates in assessable patients with 3+ and 2+ HER2 overexpression were 48% and 7%, respectively. The response rates in 108 assessable patients with and without HER2 gene amplification by fluorescence in situ hybridization (FISH) analysis were 34% (95% CI, 23.9% to 45.7%) and 7% (95% CI, 0.8% to 22.8%), respectively. Seventeen (57%) of 30 patients with an objective response and 22 (51%) of 43 patients with clinical benefit had not experienced disease progression at follow-up at 12 months or later. The most common treatment-related adverse events were chills (25% of patients), asthenia (23%), fever (22%), pain (18%), and nausea (14%). Cardiac dysfunction occurred in two patients (2%); both had histories of cardiac disease and did not require additional intervention after discontinuation of trastuzumab. There was no clear evidence of a dose-response relationship for response, survival, or adverse events. CONCLUSION Single-agent trastuzumab is active and well tolerated as first-line treatment of women with metastatic breast cancer with HER2 3+ overexpression by IHC or gene amplification by FISH.
- Published
- 2023
- Full Text
- View/download PDF
5. Phase II Study of Palbociclib (PD-0332991) in CCND1, 2, or 3 Amplification: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol Z1B
- Author
-
Amy S. Clark, Fangxin Hong, Richard S. Finn, Angela M. DeMichele, Edith P. Mitchell, James Zwiebel, Fernanda I. Arnaldez, Robert J. Gray, Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David Patton, P. Mickey Williams, Stanley R. Hamilton, Mehmet S. Copur, Samer S. Kasbari, Ravneet Thind, Barbara A. Conley, Carlos L. Arteaga, Peter J. O'Dwyer, Lyndsay N. Harris, Alice P. Chen, and Keith T. Flaherty
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Cyclin D/CDK4/6 is critical in controlling the G1 to S checkpoint. CCND, the gene encoding cyclin D, is known to be amplified in a variety of solid tumors. Palbociclib is an oral CDK4/6 inhibitor, approved in advanced breast cancer in combination with endocrine therapy. We explored the efficacy of palbociclib in patients with nonbreast solid tumors containing an amplification in CCND1, 2, or 3. Patients and Methods: Patients with tumors containing a CCND1, 2, or 3 amplification and expression of the retinoblastoma protein were assigned to subprotocol Z1B and received palbociclib 125 mg once daily for 21 days of a 28-day cycle. Tumor response was assessed every two cycles. Results: Forty patients were assigned to subprotocol Z1B; 4 patients had outside assays identifying the CCND1, 2, or 3 amplification and were not confirmed centrally; 3 were ineligible and 2 were not treated (1 untreated patient was also ineligible), leaving 32 evaluable patients for this analysis. There were no partial responses; 12 patients (37.5%) had stable disease as best response. There were seven deaths on study, all during cycle 1 and attributable to disease progression. Median progression-free survival was 1.8 months. The most common toxicities were leukopenia (n = 21, 55%) and neutropenia (n = 19, 50%); neutropenia was the most common grade 3/4 event (n = 12, 32%). Conclusions: Palbociclib was not effective at treating nonbreast solid tumors with a CCND1, 2, or 3 amplification in this cohort. These data do not support further investigation of single-agent palbociclib in tumors with CCND1, 2, or 3 amplification.
- Published
- 2023
- Full Text
- View/download PDF
6. National Cancer Institute Combination Therapy Platform Trial with Molecular Analysis for Therapy Choice (ComboMATCH)
- Author
-
Funda Meric-Bernstam, James M. Ford, Peter J. O'Dwyer, Geoffrey I. Shapiro, Lisa M. McShane, Boris Freidlin, Roisin E. O'Cearbhaill, Suzanne George, Julia Glade-Bender, Gary H. Lyman, James V. Tricoli, David Patton, Stanley R. Hamilton, Robert J. Gray, Douglas S. Hawkins, Bhanumati Ramineni, Keith T. Flaherty, Petros Grivas, Timothy A. Yap, Jordan Berlin, James H. Doroshow, Lyndsay N. Harris, and Jeffrey A. Moscow
- Subjects
Cancer Research ,Oncology - Abstract
Over the past decade, multiple trials, including the precision medicine trial National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH, EAY131, NCT02465060) have sought to determine if treating cancer based on specific genomic alterations is effective, irrespective of the cancer histology. Although many therapies are now approved for the treatment of cancers harboring specific genomic alterations, most patients do not respond to therapies targeting a single alteration. Further, when antitumor responses do occur, they are often not durable due to the development of drug resistance. Therefore, there is a great need to identify rational combination therapies that may be more effective. To address this need, the NCI and National Clinical Trials Network have developed NCI-ComboMATCH, the successor to NCI-MATCH. Like the original trial, NCI-ComboMATCH is a signal-seeking study. The goal of ComboMATCH is to overcome drug resistance to single-agent therapy and/or utilize novel synergies to increase efficacy by developing genomically-directed combination therapies, supported by strong preclinical in vivo evidence. Although NCI-MATCH was mainly comprised of multiple single-arm studies, NCI-ComboMATCH tests combination therapy, evaluating both combination of targeted agents as well as combinations of targeted therapy with chemotherapy. Although NCI-MATCH was histology agnostic with selected tumor exclusions, ComboMATCH has histology-specific and histology-agnostic arms. Although NCI-MATCH consisted of single-arm studies, ComboMATCH utilizes single-arm as well as randomized designs. NCI-MATCH had a separate, parallel Pediatric MATCH trial, whereas ComboMATCH will include children within the same trial. We present rationale, scientific principles, study design, and logistics supporting the ComboMATCH study.
- Published
- 2023
- Full Text
- View/download PDF
7. Phase II Study of Afatinib in Patients With Tumors With Human Epidermal Growth Factor Receptor 2-Activating Mutations: Results From the National Cancer Institute-Molecular Analysis for Therapy Choice ECOG-ACRIN Trial (EAY131) Subprotocol EAY131-B
- Author
-
Philippe L. Bedard, Shuli Li, Kari B. Wisinski, Eddy S. Yang, Sewanti A. Limaye, Edith P. Mitchell, James A. Zwiebel, Jeffrey A. Moscow, Robert J. Gray, Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David R. Patton, P. Mickey Williams, Stanley R. Hamilton, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, and Keith T. Flaherty
- Subjects
Diarrhea ,Male ,Cancer Research ,Receptor, ErbB-2 ,Breast Neoplasms ,Stroke Volume ,Middle Aged ,Afatinib ,National Cancer Institute (U.S.) ,United States ,Ventricular Function, Left ,Oncology ,Receptors, Estrogen ,Mutation ,Quinazolines ,Humans ,Female ,In Situ Hybridization, Fluorescence - Abstract
PURPOSE National Cancer Institute–Molecular Analysis for Therapy Choice is a multicohort trial that assigns patients with advanced cancers to targeted therapies on the basis of central tumor genomic testing. Arm B evaluated afatinib, an ErbB family tyrosine kinase inhibitor, in patients with ERBB2-activating mutations. METHODS Eligible patients had selected ERBB2 single-nucleotide variants or insertions/deletions detected by the National Cancer Institute–Molecular Analysis for Therapy Choice next-generation sequencing assay. Patients had performance status ≤ 1, left ventricular ejection fraction > 50%, grade ≤ 1 diarrhea, and no prior human epidermal growth factor receptor 2 (HER2) therapy. Patients received afatinib 40 mg once daily in 28-day cycles. The primary end point was objective response rate (ORR). Secondary end points were 6-month progression-free survival, overall survival, toxicity, and molecular correlates. RESULTS A total of 59 patients were assigned and 40 were enrolled. The median age was 62 years, 78% were female, 68% had performance status = 1, and 58% had received > 3 prior therapies. The confirmed ORR was 2.7% (n = 1 of 37; 90% CI, 0.14 to 12.2), and 6-month progression-free survival was 12.0% (90% CI, 5.6 to 25.8). A confirmed partial response occurred in a patient with adenocarcinoma of extra-mammary Paget disease of skin who progressed after cycle 6. Two unconfirmed partial responses were observed (low-grade serous gynecological tract and estrogen receptor–positive/HER2-negative immunohistochemistry breast ductal carcinoma). Of 12 patients with breast cancer, 1 additional patient with lobular carcinoma (estrogen receptor–positive/HER2 fluorescent in situ hybridization) had a 51% reduction in target lesions but progressed because of a new lesion at cycle 6. The most common (> 20%) treatment-related adverse events were diarrhea (68%), mucositis (43%), fatigue (40%), acneiform rash (30%), dehydration (27%), vomiting (27%), nausea (27%), anemia (27%), and anorexia (22%). Four patients (11%) discontinued because of adverse events. CONCLUSION Although afatinib did not meet the prespecified threshold for antitumor activity in this heavily pretreated cohort, the response in a rare tumor type is notable. The safety profile of afatinib was consistent with prior studies.
- Published
- 2023
8. Phase II Study of Copanlisib in Patients With Tumors With PIK3CA Mutations: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol Z1F
- Author
-
Senthil Damodaran, Fengmin Zhao, Dustin A. Deming, Edith P. Mitchell, John J. Wright, Robert J. Gray, Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David R. Patton, P. Mickey Williams, Stanley R. Hamilton, Jennifer M. Suga, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, and Keith T. Flaherty
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE Activating mutations in PIK3CA are observed across multiple tumor types. The NCI-MATCH (EAY131) is a tumor-agnostic platform trial that enrolls patients to targeted therapies on the basis of matching genomic alterations. Arm Z1F evaluated copanlisib, an α and δ isoform–specific phosphoinositide 3-kinase (PI3K) inhibitor, in patients with PIK3CA mutations (with or without PTEN loss). PATIENTS AND METHODS Patients received copanlisib (60 mg intravenous) once weekly on days 1, 8, and 15 in 28-day cycles until progression or toxicity. Patients with KRAS mutations, human epidermal growth factor receptor 2–positive breast cancers, and lymphomas were excluded. The primary end point was centrally assessed objective response rate (ORR); secondary end points included progression-free survival, 6-month progression-free survival, and overall survival. RESULTS Thirty-five patients were enrolled, and 25 patients were included in the primary efficacy analysis as prespecified in the Protocol. Multiple histologies were enrolled, with gynecologic (n = 6) and gastrointestinal (n = 6) being the most common. Sixty-eight percent of patients had ≥ 3 lines of prior therapy. The ORR was 16% (4 of 25, 90% CI, 6 to 33) with P = .0341 against a null rate of 5%. The most common reason for protocol discontinuation was disease progression (n = 17, 68%). Grade 3/4 toxicities observed were consistent with reported toxicities for PI3K pathway inhibition. Sixteen patients (53%) had grade 3 toxicities, and one patient (3%) had grade 4 toxicity (CTCAE v5.0). Most common toxicities include hyperglycemia (n = 19), fatigue (n = 12), diarrhea (n = 11), hypertension (n = 10), and nausea (n = 10). CONCLUSION The study met its primary end point with an ORR of 16% ( P = .0341) with copanlisib showing clinical activity in select tumors with PIK3CA mutation in the refractory setting.
- Published
- 2022
- Full Text
- View/download PDF
9. Phase II Study of Copanlisib in Patients With Tumors With
- Author
-
Senthil, Damodaran, Fengmin, Zhao, Dustin A, Deming, Edith P, Mitchell, John J, Wright, Robert J, Gray, Victoria, Wang, Lisa M, McShane, Larry V, Rubinstein, David R, Patton, P Mickey, Williams, Stanley R, Hamilton, Jennifer M, Suga, Barbara A, Conley, Carlos L, Arteaga, Lyndsay N, Harris, Peter J, O'Dwyer, Alice P, Chen, and Keith T, Flaherty
- Subjects
Phosphatidylinositol 3-Kinases ,Pyrimidines ,Class I Phosphatidylinositol 3-Kinases ,Quinazolines ,Humans ,Breast Neoplasms ,Female ,ORIGINAL REPORTS ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Activating mutations in PIK3CA are observed across multiple tumor types. The NCI-MATCH (EAY131) is a tumor-agnostic platform trial that enrolls patients to targeted therapies on the basis of matching genomic alterations. Arm Z1F evaluated copanlisib, an α and δ isoform–specific phosphoinositide 3-kinase (PI3K) inhibitor, in patients with PIK3CA mutations (with or without PTEN loss). PATIENTS AND METHODS: Patients received copanlisib (60 mg intravenous) once weekly on days 1, 8, and 15 in 28-day cycles until progression or toxicity. Patients with KRAS mutations, human epidermal growth factor receptor 2–positive breast cancers, and lymphomas were excluded. The primary end point was centrally assessed objective response rate (ORR); secondary end points included progression-free survival, 6-month progression-free survival, and overall survival. RESULTS: Thirty-five patients were enrolled, and 25 patients were included in the primary efficacy analysis as prespecified in the Protocol. Multiple histologies were enrolled, with gynecologic (n = 6) and gastrointestinal (n = 6) being the most common. Sixty-eight percent of patients had ≥ 3 lines of prior therapy. The ORR was 16% (4 of 25, 90% CI, 6 to 33) with P = .0341 against a null rate of 5%. The most common reason for protocol discontinuation was disease progression (n = 17, 68%). Grade 3/4 toxicities observed were consistent with reported toxicities for PI3K pathway inhibition. Sixteen patients (53%) had grade 3 toxicities, and one patient (3%) had grade 4 toxicity (CTCAE v5.0). Most common toxicities include hyperglycemia (n = 19), fatigue (n = 12), diarrhea (n = 11), hypertension (n = 10), and nausea (n = 10). CONCLUSION: The study met its primary end point with an ORR of 16% (P = .0341) with copanlisib showing clinical activity in select tumors with PIK3CA mutation in the refractory setting.
- Published
- 2023
10. Data from National Cancer Institute Combination Therapy Platform Trial with Molecular Analysis for Therapy Choice (ComboMATCH)
- Author
-
Jeffrey A. Moscow, Lyndsay N. Harris, James H. Doroshow, Jordan Berlin, Timothy A. Yap, Petros Grivas, Keith T. Flaherty, Bhanumati Ramineni, Douglas S. Hawkins, Robert J. Gray, Stanley R. Hamilton, David Patton, James V. Tricoli, Gary H. Lyman, Julia Glade-Bender, Suzanne George, Roisin E. O'Cearbhaill, Boris Freidlin, Lisa M. McShane, Geoffrey I. Shapiro, Peter J. O'Dwyer, James M. Ford, and Funda Meric-Bernstam
- Abstract
Over the past decade, multiple trials, including the precision medicine trial National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH, EAY131, NCT02465060) have sought to determine if treating cancer based on specific genomic alterations is effective, irrespective of the cancer histology. Although many therapies are now approved for the treatment of cancers harboring specific genomic alterations, most patients do not respond to therapies targeting a single alteration. Further, when antitumor responses do occur, they are often not durable due to the development of drug resistance. Therefore, there is a great need to identify rational combination therapies that may be more effective. To address this need, the NCI and National Clinical Trials Network have developed NCI-ComboMATCH, the successor to NCI-MATCH. Like the original trial, NCI-ComboMATCH is a signal-seeking study. The goal of ComboMATCH is to overcome drug resistance to single-agent therapy and/or utilize novel synergies to increase efficacy by developing genomically-directed combination therapies, supported by strong preclinical in vivo evidence. Although NCI-MATCH was mainly comprised of multiple single-arm studies, NCI-ComboMATCH tests combination therapy, evaluating both combination of targeted agents as well as combinations of targeted therapy with chemotherapy. Although NCI-MATCH was histology agnostic with selected tumor exclusions, ComboMATCH has histology-specific and histology-agnostic arms. Although NCI-MATCH consisted of single-arm studies, ComboMATCH utilizes single-arm as well as randomized designs. NCI-MATCH had a separate, parallel Pediatric MATCH trial, whereas ComboMATCH will include children within the same trial. We present rationale, scientific principles, study design, and logistics supporting the ComboMATCH study.
- Published
- 2023
- Full Text
- View/download PDF
11. supplementary table TS1 from National Cancer Institute Combination Therapy Platform Trial with Molecular Analysis for Therapy Choice (ComboMATCH)
- Author
-
Jeffrey A. Moscow, Lyndsay N. Harris, James H. Doroshow, Jordan Berlin, Timothy A. Yap, Petros Grivas, Keith T. Flaherty, Bhanumati Ramineni, Douglas S. Hawkins, Robert J. Gray, Stanley R. Hamilton, David Patton, James V. Tricoli, Gary H. Lyman, Julia Glade-Bender, Suzanne George, Roisin E. O'Cearbhaill, Boris Freidlin, Lisa M. McShane, Geoffrey I. Shapiro, Peter J. O'Dwyer, James M. Ford, and Funda Meric-Bernstam
- Abstract
supplementary table TS1
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table S1 from Phase II Study of Palbociclib (PD-0332991) in CCND1, 2, or 3 Amplification: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol Z1B
- Author
-
Keith T. Flaherty, Alice P. Chen, Lyndsay N. Harris, Peter J. O'Dwyer, Carlos L. Arteaga, Barbara A. Conley, Ravneet Thind, Samer S. Kasbari, Mehmet S. Copur, Stanley R. Hamilton, P. Mickey Williams, David Patton, Larry V. Rubinstein, Lisa M. McShane, Victoria Wang, Robert J. Gray, Fernanda I. Arnaldez, James Zwiebel, Edith P. Mitchell, Angela M. DeMichele, Richard S. Finn, Fangxin Hong, and Amy S. Clark
- Abstract
Representativeness of Study Participants
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Phase II Study of Palbociclib (PD-0332991) in CCND1, 2, or 3 Amplification: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol Z1B
- Author
-
Keith T. Flaherty, Alice P. Chen, Lyndsay N. Harris, Peter J. O'Dwyer, Carlos L. Arteaga, Barbara A. Conley, Ravneet Thind, Samer S. Kasbari, Mehmet S. Copur, Stanley R. Hamilton, P. Mickey Williams, David Patton, Larry V. Rubinstein, Lisa M. McShane, Victoria Wang, Robert J. Gray, Fernanda I. Arnaldez, James Zwiebel, Edith P. Mitchell, Angela M. DeMichele, Richard S. Finn, Fangxin Hong, and Amy S. Clark
- Abstract
Purpose:Cyclin D/CDK4/6 is critical in controlling the G1 to S checkpoint. CCND, the gene encoding cyclin D, is known to be amplified in a variety of solid tumors. Palbociclib is an oral CDK4/6 inhibitor, approved in advanced breast cancer in combination with endocrine therapy. We explored the efficacy of palbociclib in patients with nonbreast solid tumors containing an amplification in CCND1, 2, or 3.Patients and Methods:Patients with tumors containing a CCND1, 2, or 3 amplification and expression of the retinoblastoma protein were assigned to subprotocol Z1B and received palbociclib 125 mg once daily for 21 days of a 28-day cycle. Tumor response was assessed every two cycles.Results:Forty patients were assigned to subprotocol Z1B; 4 patients had outside assays identifying the CCND1, 2, or 3 amplification and were not confirmed centrally; 3 were ineligible and 2 were not treated (1 untreated patient was also ineligible), leaving 32 evaluable patients for this analysis. There were no partial responses; 12 patients (37.5%) had stable disease as best response. There were seven deaths on study, all during cycle 1 and attributable to disease progression. Median progression-free survival was 1.8 months. The most common toxicities were leukopenia (n = 21, 55%) and neutropenia (n = 19, 50%); neutropenia was the most common grade 3/4 event (n = 12, 32%).Conclusions:Palbociclib was not effective at treating nonbreast solid tumors with a CCND1, 2, or 3 amplification in this cohort. These data do not support further investigation of single-agent palbociclib in tumors with CCND1, 2, or 3 amplification.
- Published
- 2023
- Full Text
- View/download PDF
14. Trametinib in Patients With NF1-, GNAQ-, or GNA11-Mutant Tumors: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocols S1 and S2
- Author
-
Kari B. Wisinski, Yael Flamand, Melissa A. Wilson, Jason J. Luke, Hussein A. Tawbi, Fangxin Hong, Edith P. Mitchell, James A. Zwiebel, Helen Chen, Robert J. Gray, Shuli Li, Lisa M. McShane, Lawrence V. Rubinstein, David Patton, P. Mickey Williams, Stanley R. Hamilton, Robert J. Behrens, Kathryn P. Pennington, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, and Keith T. Flaherty
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE NCI-MATCH is a precision medicine trial using genomic testing to allocate patients with advanced malignancies to targeted treatment subprotocols. This report combines two subprotocols evaluating trametinib, a MEK1/2 inhibitor, in patients with Neurofibromatosis 1 ( NF1[S1] or GNA11/Q [S2]) altered tumors. METHODS Eligible patients had tumors with deleterious inactivating NF1 or GNA11/Q mutations by the customized Oncomine AmpliSeq panel. Prior MEK inhibitor treatment was excluded. Glioblastomas (GBMs) were permitted, including malignancies associated with germline NF1 mutations (S1 only). Trametinib was administered at 2 mg once daily over 28-day cycles until toxicity or disease progression. Primary end point was objective response rate (ORR). Secondary end points included progression-free survival (PFS) at 6 months, PFS, and overall survival. Exploratory analyses included co-occurring genomic alterations and PTEN loss. RESULTS Fifty patients were eligible and started therapy: 46 with NF1 mutations (S1) and four with GNA11 mutations (S2). In the NF1 cohort, nonsense single-nucleotide variants were identified in 29 and frameshift deletions in 17 tumors. All in S2 had nonuveal melanoma and GNA11 Q209L variant. Two partial responses (PR) were noted in S1, one patient each with advanced lung cancer and GBM for an ORR of 4.3% (90% CI, 0.8 to 13.1). One patient with melanoma in S2 had a PR (ORR, 25%; 90% CI, 1.3 to 75.1). Prolonged stable disease (SD) was also noted in five patients (four in S1 and one in S2) with additional rare histologies. Adverse events were as previously described with trametinib. Comutations in TP53 and PIK3CA were common. CONCLUSION Although these subprotocols did not meet the primary end point for ORR, significant responses or prolonged SD noted in some disease subtypes warrants further investigation.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplemental Figures and Tables from Immune Signatures Following Single Dose Trastuzumab Predict Pathologic Response to PreoperativeTrastuzumab and Chemotherapy in HER2-Positive Early Breast Cancer
- Author
-
Lyndsay N. Harris, William Sikov, Mary Anne Fenton, Maysa M. Abu-Khalaf, George Somlo, Veerle Bossuyt, Eric P. Winer, Ian E. Krop, Amad Awadallah, Shikha Parsai, David Tuck, Kristy L.S. Miskimen, Hannah Gilmore, and Vinay Varadan
- Abstract
Supplemental Figure 1. Consort Diagram 03-311. Supplemental Figure 2. Consort Diagram 211B. Supplemental Figure 3. Subtype-specific changes in Immune Index upon brief-exposure to nab-paclitaxel in the 211B trial. Supplemental Figure 4. Evaluation of the Immune Index upon brief-exposure to trastuzumab in the 03-311 and 211B trials according to ER/PR status. Supplemental Figure 5. Evaluation of the Immune Index upon brief-exposure to nab-paclitaxel in the 211B trial according to ER/PR status. Supplemental Figure 6. Correlation between Immune Index and tumor content. Supplemental Figure 7. Association of CD4+ Th1 and Treg signatures with response. Supplemental Table 1. Immune Index genes discriminative of response at the post-exposure timepoint in the discovery (03-311) and validation (211B) datasets. Supplemental Table 2. Immune cell subset signatures discriminative of response at baseline and post-exposure timepoints in the discovery (03-311) and validation (211B) datasets. Supplemental Table 3. CD8+ T-cell cytolytic activity genes discriminative of response at the post-exposure timepoint in the discovery (03-311) and validation (211B) datasets.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary eTables from Integrated Analysis of RNA and DNA from the Phase III Trial CALGB 40601 Identifies Predictors of Response to Trastuzumab-Based Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer
- Author
-
Charles M. Perou, Lisa A. Carey, Clifford A. Hudis, Eric P. Winer, Elaine R. Mardis, Donald A. Berry, Lyndsay N. Harris, Ian E. Krop, Chau T. Dang, Sara Tolaney, Lynn N. Henry, Patricia A. Spears, Matthew G. Soloway, Brandelyn N. Pitcher, Terry M. Hyslop, Yan Li, Zhiyuan Hu, Katherine A. Hoadley, Joel S. Parker, Cheng Fan, and Maki Tanioka
- Abstract
Supplementary eTables 1-12
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Tables 1-4 from Trametinib Activity in Patients with Solid Tumors and Lymphomas Harboring BRAF Non-V600 Mutations or Fusions: Results from NCI-MATCH (EAY131)
- Author
-
Keith T. Flaherty, Alice P. Chen, Peter J. O'Dwyer, Lyndsay N. Harris, Carlos L. Arteaga, Barbara A. Conley, Stanly R. Hamilton, P. Mickey Williams, David Patton, Larry V. Rubinstein, Lisa M. McShane, Shuli Li, Robert J. Gray, Helen X. Chen, John J. Wright, Edith P. Mitchell, Gregory J. Riely, Marcus Noel, Fengmin Zhao, and Douglas B. Johnson
- Abstract
1) Eligible mutations/fusions, 2) identified mutations/fusions, 3) Treatment-related toxicities, 4) Number of treatment cycles and reasons for discontinuation
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Legend from Trametinib Activity in Patients with Solid Tumors and Lymphomas Harboring BRAF Non-V600 Mutations or Fusions: Results from NCI-MATCH (EAY131)
- Author
-
Keith T. Flaherty, Alice P. Chen, Peter J. O'Dwyer, Lyndsay N. Harris, Carlos L. Arteaga, Barbara A. Conley, Stanly R. Hamilton, P. Mickey Williams, David Patton, Larry V. Rubinstein, Lisa M. McShane, Shuli Li, Robert J. Gray, Helen X. Chen, John J. Wright, Edith P. Mitchell, Gregory J. Riely, Marcus Noel, Fengmin Zhao, and Douglas B. Johnson
- Abstract
Supplementary Legend
- Published
- 2023
- Full Text
- View/download PDF
19. Data from Integrated Analysis of RNA and DNA from the Phase III Trial CALGB 40601 Identifies Predictors of Response to Trastuzumab-Based Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer
- Author
-
Charles M. Perou, Lisa A. Carey, Clifford A. Hudis, Eric P. Winer, Elaine R. Mardis, Donald A. Berry, Lyndsay N. Harris, Ian E. Krop, Chau T. Dang, Sara Tolaney, Lynn N. Henry, Patricia A. Spears, Matthew G. Soloway, Brandelyn N. Pitcher, Terry M. Hyslop, Yan Li, Zhiyuan Hu, Katherine A. Hoadley, Joel S. Parker, Cheng Fan, and Maki Tanioka
- Abstract
Purpose: Response to a complex trastuzumab-based regimen is affected by multiple features of the tumor and its microenvironment. Developing a predictive algorithm is key to optimizing HER2-targeting therapy.Experimental Design: We analyzed 137 pretreatment tumors with mRNA-seq and DNA exome sequencing from CALGB 40601, a neoadjuvant phase III trial of paclitaxel plus trastuzumab with or without lapatinib in stage II to III HER2-positive breast cancer. We adopted an Elastic Net regularized regression approach that controls for covarying features within high-dimensional data. First, we applied 517 known gene expression signatures to develop an Elastic Net model to predict pCR, which we validated on 143 samples from four independent trials. Next, we performed integrative analyses incorporating clinicopathologic information with somatic mutation status, DNA copy number alterations (CNA), and gene signatures.Results: The Elastic Net model using only gene signatures predicted pCR in the validation sets (AUC = 0.76). Integrative analyses showed that models containing gene signatures, clinical features, and DNA information were better pCR predictors than models containing a single data type. Frequently selected variables from the multiplatform models included amplifications of chromosome 6p, TP53 mutation, HER2-enriched subtype, and immune signatures. Variables predicting resistance included Luminal/ER+ features.Conclusions: Models using RNA only, as well as integrated RNA and DNA models, can predict pCR with improved accuracy over clinical variables. Somatic DNA alterations (mutation, CNAs), tumor molecular subtype (HER2E, Luminal), and the microenvironment (immune cells) were independent predictors of response to trastuzumab and paclitaxel-based regimens. This highlights the complexity of predicting response in HER2-positive breast cancer. Clin Cancer Res; 24(21); 5292–304. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure 2 from Impact of c-MYC Protein Expression on Outcome of Patients with Early-Stage HER2+ Breast Cancer Treated with Adjuvant Trastuzumab NCCTG (Alliance) N9831
- Author
-
Edith A. Perez, Wilma L. Lingle, James N. Ingle, Lyndsay N. Harris, Julie Gralow, Leila A. Kutteh, Peter A. Kaufman, George W. Sledge, Silvana Martino, Nancy E. Davidson, Ann E. McCullough, Beiyun Chen, Darren Riehle, Robert B. Jenkins, Karla Ballman, Kathleen Tenner, Xochiquetzal J. Geiger, Monica M. Reinholz, and Amylou C. Dueck
- Abstract
Supplementary Figure 2 - PDF file 9846K, Patient Flow Diagram for N9831 MYC IHC Analysis
- Published
- 2023
- Full Text
- View/download PDF
21. Supplemental Table from Differential Outcomes in Codon 12/13 and Codon 61 NRAS-Mutated Cancers in the Phase II NCI-MATCH Trial of Binimetinib in Patients with NRAS-Mutated Tumors
- Author
-
Keith T. Flaherty, Alice P. Chen, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Andrew J. Aguirre, Barbara A. Conley, Stanley R. Hamilton, P. Mickey Williams, Melissa S. Dillmon, Funda Meric-Bernstam, David R. Patton, Larry V. Rubinstein, Lisa M. McShane, Robert J. Gray, Shuli Li, Helen X. Chen, Kevin S. Kapner, James A. Zwiebel, Edith P. Mitchell, E. Scott Kopetz, Rebecca S. Heist, Victoria Wang, and James M. Cleary
- Abstract
Supplemental Table
- Published
- 2023
- Full Text
- View/download PDF
22. Data from Impact of c-MYC Protein Expression on Outcome of Patients with Early-Stage HER2+ Breast Cancer Treated with Adjuvant Trastuzumab NCCTG (Alliance) N9831
- Author
-
Edith A. Perez, Wilma L. Lingle, James N. Ingle, Lyndsay N. Harris, Julie Gralow, Leila A. Kutteh, Peter A. Kaufman, George W. Sledge, Silvana Martino, Nancy E. Davidson, Ann E. McCullough, Beiyun Chen, Darren Riehle, Robert B. Jenkins, Karla Ballman, Kathleen Tenner, Xochiquetzal J. Geiger, Monica M. Reinholz, and Amylou C. Dueck
- Abstract
Purpose: This study investigated the association between tumor MYC protein expression and disease-free survival (DFS) of patients randomized to receive chemotherapy alone (Arm A) or chemotherapy with sequential (Arm B) or concurrent trastuzumab (Arm C) in the N9831 (Alliance) adjuvant HER2+ trastuzumab breast cancer trial.Experimental Design: This analysis included 1,736 patients randomized to Arms A, B, and C on N9831. Nuclear MYC protein expression was determined in tissue microarray sections containing three biopsies per patient or whole tissue sections using standard immunohistochemistry (clone 9E10). A tumor was considered positive for MYC protein overexpression (MYC+) if the nuclear 3+ staining percentage was more than 30%.Results: Five hundred and seventy-four (33%) tumors were MYC+. MYC+ was associated with hormone receptor positivity (χ2, P = 0.006), tumors 2 cm or more (χ2, P = 0.02), and a higher rate of nodal positivity (χ2, P < 0.001). HRs for DFS (median follow-up: 6.1 years) for Arm C versus A were 0.52 (P = 0.006) and 0.65 (P = 0.006) for patients with MYC+ and MYC− tumors, respectively (Pinteraction = 0.40). For Arm B versus A, HRs for patients with MYC+ and MYC− tumors were 0.79 (P = 0.21) and 0.74 (P = 0.04), respectively (Pinteraction = 0.71). For Arm C versus B, HRs for patients with MYC+ and MYC− tumors were 0.56 (P = 0.02) and 0.89 (P = 0.49), respectively (Pinteraction = 0.17).Conclusions: Our data do not support an impact of tumor MYC protein expression on differential benefit from adjuvant trastuzumab. Clin Cancer Res; 19(20); 5798–807. ©2013 AACR.
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Differential Outcomes in Codon 12/13 and Codon 61 NRAS-Mutated Cancers in the Phase II NCI-MATCH Trial of Binimetinib in Patients with NRAS-Mutated Tumors
- Author
-
Keith T. Flaherty, Alice P. Chen, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Andrew J. Aguirre, Barbara A. Conley, Stanley R. Hamilton, P. Mickey Williams, Melissa S. Dillmon, Funda Meric-Bernstam, David R. Patton, Larry V. Rubinstein, Lisa M. McShane, Robert J. Gray, Shuli Li, Helen X. Chen, Kevin S. Kapner, James A. Zwiebel, Edith P. Mitchell, E. Scott Kopetz, Rebecca S. Heist, Victoria Wang, and James M. Cleary
- Abstract
Purpose:Preclinical and clinical data suggest that downstream inhibition with an MEK inhibitor, such as binimetinib, might be efficacious for NRAS-mutated cancers.Patients and Methods:Patients enrolled in the NCI-MATCH trial master protocol underwent tumor biopsy and molecular profiling by targeted next-generation sequencing. Patients with NRAS-mutated tumors, except melanoma, were enrolled in subprotocol Z1A, a single-arm study evaluating binimetinib 45 mg twice daily. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS) and overall survival (OS). A post hoc analysis examined the association of NRAS mutation type with outcome.Results:In total, 47 eligible patients with a refractory solid tumor harboring a codon 12, 13, or 61 NRAS mutation were treated. Observed toxicity was moderate, and 30% of patients discontinued treatment because of binimetinib-associated toxicity. The ORR was 2.1% (1/47 patients). A patient with malignant ameloblastoma harboring a codon 61 NRAS mutation achieved a durable partial response (PR). A patient with NRAS codon 61–mutated colorectal cancer had an unconfirmed PR, and two other patients with NRAS codon 61–mutated colorectal had stable disease for at least 12 months. In an exploratory analysis, patients with colorectal cancer bearing a NRAS codon 61 mutation (n = 8) had a significantly longer OS (P = 0.03) and PFS (P = 0.007) than those with codon 12 or 13 mutations (n = 16).Conclusions:Single-agent binimetinib did not show promising efficacy in NRAS-mutated cancers. The observation of increased OS and PFS in patients with codon 61 NRAS-mutated colorectal cancer merits further investigation.
- Published
- 2023
- Full Text
- View/download PDF
24. Data from Trametinib Activity in Patients with Solid Tumors and Lymphomas Harboring BRAF Non-V600 Mutations or Fusions: Results from NCI-MATCH (EAY131)
- Author
-
Keith T. Flaherty, Alice P. Chen, Peter J. O'Dwyer, Lyndsay N. Harris, Carlos L. Arteaga, Barbara A. Conley, Stanly R. Hamilton, P. Mickey Williams, David Patton, Larry V. Rubinstein, Lisa M. McShane, Shuli Li, Robert J. Gray, Helen X. Chen, John J. Wright, Edith P. Mitchell, Gregory J. Riely, Marcus Noel, Fengmin Zhao, and Douglas B. Johnson
- Abstract
Purpose:Substantial preclinical evidence and case reports suggest that MEK inhibition is an active approach in tumors with BRAF mutations outside the V600 locus, and in BRAF fusions. Thus, Subprotocol R of the NCI-MATCH study tested the MEK inhibitor trametinib in this population.Patients and Methods:The NCI-MATCH study performed genomic profiling on tumor samples from patients with solid tumors and lymphomas progressing on standard therapies or with no standard treatments. Patients with prespecified fusions and non-V600 mutations in BRAF were assigned to Subprotocol R using the NCI-MATCHBOX algorithm. The primary endpoint was objective response rate (ORR).Results:Among 50 patients assigned, 32 were eligible and received therapy with trametinib. Of these, 1 had a BRAF fusion and 31 had BRAF mutations (13 and 19 with class 2 and 3 mutations, respectively). There were no complete responses; 1 patient (3%) had a confirmed partial response (patient with breast ductal adenocarcinoma with BRAF G469E mutation) and 10 patients had stable disease as best response (clinical benefit rate 34%). Median progression-free survival (PFS) was 1.8 months, and median overall survival was 5.7 months. Exploratory subgroup analyses showed that patients with colorectal adenocarcinoma (n = 8) had particularly poor PFS. No new toxicity signals were identified.Conclusions:Trametinib did not show promising clinical activity in patients with tumors harboring non-V600 BRAF mutations, and the subprotocol did not meet its primary endpoint.
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Data from Trametinib Activity in Patients with Solid Tumors and Lymphomas Harboring BRAF Non-V600 Mutations or Fusions: Results from NCI-MATCH (EAY131)
- Author
-
Keith T. Flaherty, Alice P. Chen, Peter J. O'Dwyer, Lyndsay N. Harris, Carlos L. Arteaga, Barbara A. Conley, Stanly R. Hamilton, P. Mickey Williams, David Patton, Larry V. Rubinstein, Lisa M. McShane, Shuli Li, Robert J. Gray, Helen X. Chen, John J. Wright, Edith P. Mitchell, Gregory J. Riely, Marcus Noel, Fengmin Zhao, and Douglas B. Johnson
- Abstract
Supplementary Figure 2: "Swimmer's plot as in Figure 2B also demonstrating mutation locations in individual patients, showing treatment duration for all 32 evaluable patients and their occurrence of response (*), disease progression (+) and death (#)
- Published
- 2023
- Full Text
- View/download PDF
26. Supplemental Figures from Differential Outcomes in Codon 12/13 and Codon 61 NRAS-Mutated Cancers in the Phase II NCI-MATCH Trial of Binimetinib in Patients with NRAS-Mutated Tumors
- Author
-
Keith T. Flaherty, Alice P. Chen, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Andrew J. Aguirre, Barbara A. Conley, Stanley R. Hamilton, P. Mickey Williams, Melissa S. Dillmon, Funda Meric-Bernstam, David R. Patton, Larry V. Rubinstein, Lisa M. McShane, Robert J. Gray, Shuli Li, Helen X. Chen, Kevin S. Kapner, James A. Zwiebel, Edith P. Mitchell, E. Scott Kopetz, Rebecca S. Heist, Victoria Wang, and James M. Cleary
- Abstract
Supplemental Figures
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Tables 1-5 from Impact of c-MYC Protein Expression on Outcome of Patients with Early-Stage HER2+ Breast Cancer Treated with Adjuvant Trastuzumab NCCTG (Alliance) N9831
- Author
-
Edith A. Perez, Wilma L. Lingle, James N. Ingle, Lyndsay N. Harris, Julie Gralow, Leila A. Kutteh, Peter A. Kaufman, George W. Sledge, Silvana Martino, Nancy E. Davidson, Ann E. McCullough, Beiyun Chen, Darren Riehle, Robert B. Jenkins, Karla Ballman, Kathleen Tenner, Xochiquetzal J. Geiger, Monica M. Reinholz, and Amylou C. Dueck
- Abstract
Supplementary Tables 1-5 - PDF file 91K, Supplementary Table 1. Patient Characteristics by IHC cMYC Cohort vs Non-Cohort; Supplementary Table 2. Correlation of MYC Nuclear Staining with HER2 FISH - Overall; Supplementary Table 3. Correlation of MYC Nuclear Staining with HER2 FISH - Arm A; Supplementary Data Table 4. Correlation of MYC Nuclear Staining with HER2 FISH - Arm B; Supplementary Table 5. Correlation of MYC Nuclear Staining with HER2 FISH - Arm C
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Figure Legend from Differential Outcomes in Codon 12/13 and Codon 61 NRAS-Mutated Cancers in the Phase II NCI-MATCH Trial of Binimetinib in Patients with NRAS-Mutated Tumors
- Author
-
Keith T. Flaherty, Alice P. Chen, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Andrew J. Aguirre, Barbara A. Conley, Stanley R. Hamilton, P. Mickey Williams, Melissa S. Dillmon, Funda Meric-Bernstam, David R. Patton, Larry V. Rubinstein, Lisa M. McShane, Robert J. Gray, Shuli Li, Helen X. Chen, Kevin S. Kapner, James A. Zwiebel, Edith P. Mitchell, E. Scott Kopetz, Rebecca S. Heist, Victoria Wang, and James M. Cleary
- Abstract
Supplementary Figure Legend
- Published
- 2023
- Full Text
- View/download PDF
29. Supplementary Figure 1 from Impact of c-MYC Protein Expression on Outcome of Patients with Early-Stage HER2+ Breast Cancer Treated with Adjuvant Trastuzumab NCCTG (Alliance) N9831
- Author
-
Edith A. Perez, Wilma L. Lingle, James N. Ingle, Lyndsay N. Harris, Julie Gralow, Leila A. Kutteh, Peter A. Kaufman, George W. Sledge, Silvana Martino, Nancy E. Davidson, Ann E. McCullough, Beiyun Chen, Darren Riehle, Robert B. Jenkins, Karla Ballman, Kathleen Tenner, Xochiquetzal J. Geiger, Monica M. Reinholz, and Amylou C. Dueck
- Abstract
Supplementary Figure 1 - PDF file 5863K, NCCTG N9831 Trial Incorporating Trastuzumab in Adjuvant Therapy
- Published
- 2023
- Full Text
- View/download PDF
30. Supplementary Tables S1-S3 from Predictors of Resistance to Preoperative Trastuzumab and Vinorelbine for HER2-Positive Early Breast Cancer
- Author
-
Eric P. Winer, J. Dirk Iglehart, Rebecca Gelman, Paula N. Friedman, Madhavi Kamma, Beth-Ann Lesnikoski, Harold J. Burstein, Steven E. Come, Paula D. Ryan, Dennis Sgroi, Xin Lu, Agnes Witkiewicz, Stuart J. Schnitt, Fanglei You, and Lyndsay N. Harris
- Abstract
Supplementary Tables S1-S3 from Predictors of Resistance to Preoperative Trastuzumab and Vinorelbine for HER2-Positive Early Breast Cancer
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Immune Signatures Following Single Dose Trastuzumab Predict Pathologic Response to PreoperativeTrastuzumab and Chemotherapy in HER2-Positive Early Breast Cancer
- Author
-
Lyndsay N. Harris, William Sikov, Mary Anne Fenton, Maysa M. Abu-Khalaf, George Somlo, Veerle Bossuyt, Eric P. Winer, Ian E. Krop, Amad Awadallah, Shikha Parsai, David Tuck, Kristy L.S. Miskimen, Hannah Gilmore, and Vinay Varadan
- Abstract
Purpose: Recent data suggest that intrinsic subtype and immune cell infiltration may predict response to trastuzumab-based therapy. We studied the interaction between these factors, changes in immune signatures following brief exposure to trastuzumab, and achievement of pathologic complete response (pCR) to subsequent preoperative trastuzumab and chemotherapy in HER2-positive breast cancer.Experimental Design: In patients enrolled on two multicenter trials (03-311 and 211B), tumor core biopsies were obtained at baseline and after brief exposure to single-agent trastuzumab or nab-paclitaxel. Gene expression profiles were assessed to assign PAM50 subtypes, measure immune cell activation, and were correlated with response.Results: The pCR rate was significantly higher in HER2-enriched tumors in the Discovery, 03-311 (36%, P = 0.043) dataset, as compared with other subtypes, which validated in 211B (50%, P = 0.048). Significant increases in a signature of immune cell admixture (Immune Index) were observed only following brief exposure to trastuzumab in HER2-enriched tumors (Discovery/03-311, P = 0.05; Validation/211B, P = 0.02). Increased Immune Index was predictive of response after brief exposure (03-311, P = 0.03; 211B, P = 0.04), but not at baseline, in addition to increased expression of a CD4+ follicular helper T-cell signature (03-311, P = 0.05; 211B, P = 0.04). Brief exposure to trastuzumab significantly increased gene expression of the T-cell marker PD-1 in HER2-enriched tumors (Discovery/03-311, P = 0.045) and PD-1 positivity by IHC (Validation/211B, P = 0.035).Conclusions: Correlations between pCR rates, increases in Immune Index and markers of T-cell activity following brief exposure to trastuzumab in HER2-enriched tumors provide novel insights into the interaction between tumor biology, antitumor immunity, and response to treatment, and suggest potential clinically useful biomarkers in HER2+ breast cancers. Clin Cancer Res; 22(13); 3249–59. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figures from Integrated Analysis of RNA and DNA from the Phase III Trial CALGB 40601 Identifies Predictors of Response to Trastuzumab-Based Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer
- Author
-
Charles M. Perou, Lisa A. Carey, Clifford A. Hudis, Eric P. Winer, Elaine R. Mardis, Donald A. Berry, Lyndsay N. Harris, Ian E. Krop, Chau T. Dang, Sara Tolaney, Lynn N. Henry, Patricia A. Spears, Matthew G. Soloway, Brandelyn N. Pitcher, Terry M. Hyslop, Yan Li, Zhiyuan Hu, Katherine A. Hoadley, Joel S. Parker, Cheng Fan, and Maki Tanioka
- Abstract
Supplementary Figures 1-5
- Published
- 2023
- Full Text
- View/download PDF
33. Data from Predictors of Resistance to Preoperative Trastuzumab and Vinorelbine for HER2-Positive Early Breast Cancer
- Author
-
Eric P. Winer, J. Dirk Iglehart, Rebecca Gelman, Paula N. Friedman, Madhavi Kamma, Beth-Ann Lesnikoski, Harold J. Burstein, Steven E. Come, Paula D. Ryan, Dennis Sgroi, Xin Lu, Agnes Witkiewicz, Stuart J. Schnitt, Fanglei You, and Lyndsay N. Harris
- Abstract
Purpose: To assess pathologic complete response (pCR), clinical response, feasibility, safety, and potential predictors of response to preoperative trastuzumab plus vinorelbine in patients with operable, human epidermal growth factor receptor 2 (HER2)–positive breast cancer.Experimental Design: Forty-eight patients received preoperative trastuzumab and vinorelbine weekly for 12 weeks. Single and multigene biomarker studies were done in an attempt to identify predictors of response.Results: Eight of 40 (20%) patients achieved pCR (95% confidence interval, 9-36%). Of 9 additional patients recruited for protocol-defined toxicity analysis, 8 were evaluable; 42 of 48 (88%) patients had clinical response (16 patients, clinical complete response; 26 patients, clinical partial response). T1 tumors more frequently exhibited clinical complete response (P = 0.05) and showed a trend to exhibit pCR (P = 0.07). Five (13%) patients experienced grade 1 cardiac dysfunction during preoperative treatment. Neither HER2 nor estrogen receptor status changed significantly after exposure to trastuzumab and vinorelbine. RNA profiling identified three top-level clusters by unsupervised analysis. Tumors with extremes of response [pCR (n = 3) versus nonresponse (n = 3)] fell into separate groups by hierarchical clustering. No predictive genes were identified in pCR tumors. Nonresponding tumors were more likely to be T4 stage (P = 0.02) and express basal markers (P < 0.00001), growth factors, and growth factor receptors. Insulin-like growth factor-I receptor membrane expression was associated with a lower response rate (50% versus 97%; P = 0.001).Conclusions: Preoperative trastuzumab plus vinorelbine is active and well tolerated in patients with HER2-positive, operable, stage II/III breast cancer. HER2-overexpressing tumors with a basal-like phenotype, or with expression of insulin-like growth factor-I receptor and other proteins involved in growth factor pathways, are more likely to be resistant to this regimen.
- Published
- 2023
- Full Text
- View/download PDF
34. Identifying RNAseq-based coding-noncoding co-expression interactions in breast cancer.
- Author
-
Nilanjana Banerjee, Sonia Chothani, Lyndsay N. Harris, and Nevenka Dimitrova
- Published
- 2013
- Full Text
- View/download PDF
35. Identification and characterization of gene fusions in breast cancer - A non-trivial pursuit.
- Author
-
Vinay Varadan, Vartika Agrawal, Lyndsay N. Harris, and Nevenka Dimitrova
- Published
- 2013
- Full Text
- View/download PDF
36. Statistical assessment of gene fusion detection algorithms using RNA Sequencing Data.
- Author
-
Vinay Varadan, Angel Janevski, Sitharthan Kamalakaran, Nilanjana Banerjee, Nevenka Dimitrova, and Lyndsay N. Harris
- Published
- 2012
- Full Text
- View/download PDF
37. Phase II Study of Taselisib in PIK3CA-Mutated Solid Tumors Other Than Breast and Squamous Lung Cancer: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol I
- Author
-
Ian E. Krop, Opeyemi A. Jegede, Juneko E. Grilley-Olson, Josh D. Lauring, Edith P. Mitchell, James A. Zwiebel, Robert J. Gray, Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David Patton, P. Mickey Williams, Stanley R. Hamilton, Scott A. Kono, James M. Ford, Agustin A. Garcia, Xingwei D. Sui, Robert D. Siegel, Brian M. Slomovitz, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, and Keith T. Flaherty
- Subjects
Cancer Research ,Oxazepines ,Phosphatidylinositol 3-Kinases ,Lung Neoplasms ,Oncology ,Class I Phosphatidylinositol 3-Kinases ,Carcinoma, Squamous Cell ,Imidazoles ,Humans ,ORIGINAL REPORTS ,National Cancer Institute (U.S.) ,United States - Abstract
PURPOSE PIK3CA mutations frequently contribute to oncogenesis in solid tumors. Taselisib, a potent and selective inhibitor of phosphoinositide 3-kinase, has demonstrated clinical activity in PIK3CA-mutant breast cancer. Whether PIK3CA mutations predict sensitivity to taselisib in other cancer types is unknown. National Cancer Institute–Molecular Analysis for Therapy Choice Arm EAY131-I is a single-arm, phase II study of the safety and efficacy of taselisib in patients with advanced cancers. METHODS Eligible patients had tumors with an activating PIK3CA mutation. Patients with breast or squamous cell lung carcinoma, or whose cancer had KRAS or PTEN mutations, were excluded. Patients received taselisib 4 mg, orally once daily continuously, until disease progression or unacceptable toxicity. The primary end point was objective response rate. Secondary end points included progression-free survival (PFS), 6-month PFS, overall survival (OS), and identification of predictive biomarkers. RESULTS Seventy patients were enrolled, and 61 were eligible and initiated protocol therapy. Types of PIK3CA mutations included helical 41 of 61 (67%), kinase 11 of 61 (18%), and other 9 of 61 (15%). With a median follow-up of 35.7 months, there were no complete or partial responses. Six-month PFS was 19.9% (90% CI, 12.0 to 29.3) and median PFS was 3.1 months (90% CI, 1.8 to 3.7). Six-month OS was 60.7% (90% CI, 49.6 to 70.0) and median OS was 7.2 months (90% CI, 5.9 to 10.0). Individual comutations were too heterogeneous to correlate with clinical outcome. Fatigue, diarrhea, nausea, and hyperglycemia were the most common toxicities, and most were grade 1 and 2. CONCLUSION In this study, taselisib monotherapy had very limited activity in a heterogeneous cohort of heavily pretreated cancer patients with PIK3CA-mutated tumors; the presence of a PIK3CA mutation alone does not appear to be a sufficient predictor of taselisib activity.
- Published
- 2022
38. Abstract CT160: BVD-523FB (Ulixertinib) in Patients with Tumors with BRAF Fusions, or with Non-V600E, Non-V600K BRAF Mutations: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Sub-protocol EAY131-Z1L
- Author
-
Vivek Subbiah, Fengmin Fengmin, Ragini Kudchadkar, Ryan J. Sullivan, Edith P. Mitchell, John J. Wright, Helen X. Chen, Robert J. Gray, Xin Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David Patton, P. Mickey Williams, Tilak K. Sundaresan, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, and Keith T. Flaherty
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Mutations in BRAF at codons other than V600 (non-V600) and BRAF fusions confer dependence on RAF-MEK-ERK pathway. BVD-523FB (ulixertinib) is a small molecule that potently inhibits both ERK1 and ERK2 protein kinases in the sub-nanomolar range. Based on the reports of early clinical activity in the phase 1 trial, including in non-V600 BRAF mutations, subprotocol Z1L (EAY131-Z1L) sought to investigate the clinical activity of ulixertinib in patients with tumors harboring these alterations. Methods: In this single-arm study, patients with BRAF non-V600 mutation or BRAF fusion were given ulixertinib orally at a dose of 600 mg twice daily, continuously for each 28-day cycle until progression or intolerability. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), 6-month PFS, and overall survival (OS). BRAF mutation status was determined by an analytically validated assay in a CLIA-certified laboratory for all patients. Results: From August 2019 to July 2020, 35 patients were enrolled and received protocol treatment on the trial. Among the 34 patients who were eligible, median age was 66.5; 50% were female, 88% were white, 9% black, 1% Asian. Performance status was ECOG PS 1 in 74% of patients, with remaining PS 0. Median number of prior therapies was >3.Tumor types included multiple gastrointestinal malignancies (N=16), lung cancer (N=3), and melanoma (N=3), among others. Mutations were centrally confirmed in 26 patients who were deemed analyzable per protocol. Twenty-two patients had a single nucleotide variant (SNV) in BRAF; one patient had an insertion/deletion (indel) in BRAF, and three patients harbored BRAF fusions. No patients achieved CR or PR, resulting in ORR = 0%. Stable disease was the best response in 7/26 centrally confirmed cases. Median PFS was 1.8 months (90% CI: 1.6, 2.2), 6-month PFS rate was 11% (90% CI: 4%, 22%), and median OS was 4.0 months (90% CI: 2.8, 7.4). Twenty patients (57%) had grade 3 toxicities, and one patient (3%) had grade 4 toxicity; there were no grade 5 toxicities. Most common toxicities include anemia (n=11), diarrhea (n=16), nausea (n=16), vomiting (n=11), fatigue (n=16), increased creatinine (n=12), and acneiform rash (n=14). Conclusion: BVD-523FB (ulixertinib) had no demonstrable evidence of clinical activity in this small, heavily pre-treated population of patients with tumors harboring BRAF fusions, or with non-V600E, non-V600K BRAF mutations Citation Format: Vivek Subbiah, Fengmin Fengmin, Ragini Kudchadkar, Ryan J. Sullivan, Edith P. Mitchell, John J. Wright, Helen X. Chen, Robert J. Gray, Xin Victoria Wang, Lisa M. McShane, Larry V. Rubinstein, David Patton, P. Mickey Williams, Tilak K. Sundaresan, Barbara A. Conley, Carlos L. Arteaga, Lyndsay N. Harris, Peter J. O'Dwyer, Alice P. Chen, Keith T. Flaherty. BVD-523FB (Ulixertinib) in Patients with Tumors with BRAF Fusions, or with Non-V600E, Non-V600K BRAF Mutations: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Sub-protocol EAY131-Z1L [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT160.
- Published
- 2022
- Full Text
- View/download PDF
39. Differential Outcomes in Codon 12/13 and Codon 61
- Author
-
James M, Cleary, Victoria, Wang, Rebecca S, Heist, E Scott, Kopetz, Edith P, Mitchell, James A, Zwiebel, Kevin S, Kapner, Helen X, Chen, Shuli, Li, Robert J, Gray, Lisa M, McShane, Larry V, Rubinstein, David R, Patton, Funda, Meric-Bernstam, Melissa S, Dillmon, P Mickey, Williams, Stanley R, Hamilton, Barbara A, Conley, Andrew J, Aguirre, Peter J, O'Dwyer, Lyndsay N, Harris, Carlos L, Arteaga, Alice P, Chen, and Keith T, Flaherty
- Subjects
Adult ,Aged, 80 and over ,Male ,Membrane Proteins ,Middle Aged ,Jaw Neoplasms ,Article ,GTP Phosphohydrolases ,Ameloblastoma ,Treatment Outcome ,Mutation ,Humans ,Benzimidazoles ,Female ,Codon ,Colorectal Neoplasms ,Aged - Abstract
PURPOSE: Preclinical and clinical data suggest that downstream inhibition with a MEK inhibitor, such as binimetinib, might be efficacious for NRAS-mutated cancers. PATIENTS AND METHODS: Patients enrolled in the NCI-MATCH trial master protocol underwent tumor biopsy and molecular profiling by targeted next-generation sequencing. Patients with NRAS-mutated tumors, except melanoma, were enrolled in subprotocol Z1A, a single-arm study evaluating binimetinib 45 mg twice daily. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS) and overall survival (OS). A post-hoc analysis examined the association of NRAS mutation type with outcome. RESULTS: In total, 47 eligible patients with a refractory solid tumor harboring a codon 12, 13, or 61 NRAS mutation were treated. Observed toxicity was moderate, and 30% of patients discontinued treatment because of binimetinib-associated toxicity. The ORR was 2.1% (1 of 47 patients). A malignant ameloblastoma patient harboring a codon 61 NRAS mutation achieved a durable partial response (PR). A NRAS codon 61 mutated colorectal cancer patient had an unconfirmed PR, and two other NRAS codon 61 mutated colorectal patients had stable disease for at least 12 months. In an exploratory analysis, colorectal cancer patients bearing a NRAS codon 61 mutation (n = 8) had a significantly longer OS (p = 0.03) and PFS (p = 0.007) than those with codon 12 or 13 mutations (n = 16). CONCLUSIONS: Single-agent binimetinib did not show promising efficacy in NRAS-mutated cancers. The observation of increased OS and PFS in codon 61 NRAS-mutated colorectal cancer patients merits further investigation.
- Published
- 2021
40. Dabrafenib and Trametinib in Patients With Tumors With
- Author
-
April K S, Salama, Shuli, Li, Erin R, Macrae, Jong-In, Park, Edith P, Mitchell, James A, Zwiebel, Helen X, Chen, Robert J, Gray, Lisa M, McShane, Larry V, Rubinstein, David, Patton, P Mickey, Williams, Stanley R, Hamilton, Deborah K, Armstrong, Barbara A, Conley, Carlos L, Arteaga, Lyndsay N, Harris, Peter J, O'Dwyer, Alice P, Chen, and Keith T, Flaherty
- Subjects
Adult ,Aged, 80 and over ,Male ,Proto-Oncogene Proteins B-raf ,Pyridones ,MAP Kinase Kinase 2 ,Imidazoles ,MAP Kinase Kinase 1 ,Pyrimidinones ,ORIGINAL REPORTS ,Middle Aged ,Young Adult ,Clinical Trials, Phase II as Topic ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Mutation ,Oximes ,Humans ,Female ,Aged - Abstract
PURPOSE: BRAF(V600) mutations are commonly found in melanoma and thyroid cancers and to a lesser degree in other tumor types. Subprotocol H (EAY131-H) of the NCI-MATCH platform trial sought to investigate the selective BRAF inhibitor dabrafenib and the MEK1/2 inhibitor trametinib in patients with solid tumors, lymphomas, or multiple myeloma whose tumors harbored a BRAF(V600) mutation. PATIENTS AND METHODS: EAY131-H is an open-label, single-arm study. Patients with melanoma, thyroid, or colorectal cancer were excluded; patients with non–small-cell lung cancer were later excluded in an amendment. Patients received dabrafenib 150 mg twice per day and trametinib 2 mg per day continuously until disease progression or intolerable toxicity. The primary end point was centrally assessed objective response rate (ORR); secondary end points included progression-free survival (PFS), 6-month PFS, and overall survival. RESULTS: Thirty-five patients were enrolled, and 29 were included in the primary efficacy analysis as prespecified in the protocol. Median age was 59 years, and 45% of the patients had received ≥ 3 lines of therapy. The confirmed ORR was 38% (90% CI, 22.9% to 54.9%) with P < .0001 against a null rate of 5%, and PFS was 11.4 months (90% CI, 8.4 to 16.3 months); responses were seen in 7 distinct tumor types. Seven patients had a duration of response of > 12 months, including 4 patients with a duration of response of > 24 months. An additional 8 patients had a PFS > 6 months. The median overall survival was 28.6 months. Reported adverse events were comparable to those noted in previously reported profiles of dabrafenib and trametinib. CONCLUSION: This study met its primary end point, with an ORR of 38% (P < .0001) in this mixed histology, pretreated cohort. This promising activity warrants additional investigations in BRAF(V600)-mutated tumors outside of currently approved indications.
- Published
- 2020
41. Abstract LBA003: Erdafitinib in patients with tumors harboring FGFR gene mutations or fusions: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Sub-protocol K2
- Author
-
Alain C Mita, Zihan Wei, Ingrid A Mayer, Heather Cheng, Edith P Mitchell, John J Wright, Percy Ivy, Robert J Gray, Victoria Wang, Lisa M McShane, Larry V Rubinstein, David R Patton, Mickey Williams, Stanley R Hamilton, Barbara A Conley, Carlos L Arteaga, Lyndsay N Harris, Peter J O'Dwyer, Alice P Chen, and Keith T Flaherty
- Subjects
Cancer Research ,Oncology - Abstract
Background: The NCI-MATCH precision medicine trial assigns patients (pts) with solid tumors, lymphoma, or multiple myeloma whose cancers have progressed on prior treatment to a targeted therapy based on genetic alterations identified in pre-treatment biopsies. Arm K2 (EAY131-K2) evaluated the pan-FGFR inhibitor erdafitinib (E) in pts with FGFR mutations or fusions. Patients and methods: Pts with bladder or urothelial cancers were excluded. Pts received E 8 mg PO daily (28-day cycle) until disease progression or unacceptable toxicity; dose reduction for toxicities was allowed; imaging was performed every 2 cycles. The primary endpoint was objective response rate (ORR); secondary endpoints included progression-free survival (PFS), 6-month PFS, and overall survival (OS). Results: A total of 35 pts were enrolled to this arm from 07/2018-07/2019; one was ineligible and one did not receive treatment. Nine distinct tumor histologies were represented, most common being pancreatobiliary (11), CNS (7) and gynecological tumors (5). 73% of pts were female, with median age of 59y (range 26-83y), 70% were Caucasian, and 61% of pts had received at least 3 prior therapies (range 0-22). Alterations in FGFR1, FGFR2 and FGFR3 were recorded in 6, 18, and 9 evaluable pts, respectively. 18 pt tumors had fusions and 15 had mutations in an FGFR gene. The confirmed ORR was 12% (90% CI 4%, 26%), with a median duration of response (DoR) of 7.3 months (mo), range 4.2-11.7 mo. Responses were seen in cholangiocarcinoma (2 pts), Brenner ovarian tumor and adenoid cystic carcinoma (1 pt each). Two (50%) of these 4 tumors harbored FGFR fusions and 2 FGFR mutations. 13 pts had stable disease (SD). Median PFS was 3.9 mo, and 6-mo PFS was 32.8% (90% CI 21.2%, 50.6%). Median OS was 11.0 mo. Of the 6 pts with intrahepatic cholangiocarcinoma, 2 had PR and 2 SD. The most frequent grade 3 treatment-related AEs were oral mucositis/pain (5 pts), paronychia, electrolyte disorders, and anemia/lymphopenia (2 pts each). There were no treatment-related grade 4-5 toxicities. Toxicities were reversible and manageable with E dose interruptions and/or dose reduction. Conclusions: In this pre-treated, mixed histology cohort with tumors harboring FGFR somatic alterations, E showed activity with durable responses and disease stabilizations outside of currently approved FDA indications, although the pre-specified criterion that the primary endpoint, ORR, be significantly greater than 16% was not reached. Toxicities were consistent with E safety profile. Responses were observed in tumors harboring FGFR fusions as well as in those with mutations of FGFR; further correlative analyses are planned. Citation Format: Alain C Mita, Zihan Wei, Ingrid A Mayer, Heather Cheng, Edith P Mitchell, John J Wright, Percy Ivy, Robert J Gray, Victoria Wang, Lisa M McShane, Larry V Rubinstein, David R Patton, Mickey Williams, Stanley R Hamilton, Barbara A Conley, Carlos L Arteaga, Lyndsay N Harris, Peter J O'Dwyer, Alice P Chen, Keith T Flaherty. Erdafitinib in patients with tumors harboring FGFR gene mutations or fusions: Results from the NCI-MATCH ECOG-ACRIN Trial (EAY131) Sub-protocol K2 [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2021 Oct 7-10. Philadelphia (PA): AACR; Mol Cancer Ther 2021;20(12 Suppl):Abstract nr LBA003.
- Published
- 2021
- Full Text
- View/download PDF
42. Recursive SVM feature selection and sample classification for mass-spectrometry and microarray data.
- Author
-
Xuegong Zhang, Xin Lu, Qian Shi, Xiu-qin Xu, Hon-chiu E. Leung, Lyndsay N. Harris, James D. Iglehart, Alexander Miron, Jun S. Liu, and Wing Hung Wong
- Published
- 2006
- Full Text
- View/download PDF
43. Use of Biomarkers to Guide Decisions on Systemic Therapy for Women With Metastatic Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline
- Author
-
Lyndsay N. Harris, Mark R. Somerfield, and Catherine Van Poznak
- Subjects
Oncology ,Clinical Oncology ,medicine.medical_specialty ,Oncology (nursing) ,business.industry ,Health Policy ,MEDLINE ,Guideline ,medicine.disease ,Metastatic breast cancer ,Systemic therapy ,Clinical Practice ,Internal medicine ,medicine ,business - Published
- 2015
- Full Text
- View/download PDF
44. Biomarkers for Predicting Response to Anti-HER2 Agents
- Author
-
Vinay, Varadan, Maria, Sandoval, and Lyndsay N, Harris
- Subjects
Receptor, ErbB-2 ,Patient Selection ,Antineoplastic Agents ,Breast Neoplasms ,Risk Assessment ,Treatment Outcome ,Drug Resistance, Neoplasm ,Predictive Value of Tests ,Risk Factors ,Biomarkers, Tumor ,Neoplastic Stem Cells ,Animals ,Humans ,Female ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Signal Transduction - Abstract
The HER2 receptor is amplified or overexpressed in approximately 20% of all breast cancers, but despite significant efforts of the clinical research community and a growing number of anti-HER2 agents, a significant number of patients with HER2-positive breast cancer either progress or suffer disease relapse within 5-10 years. The development of robust biomarkers that predict response to anti-HER2 agents is therefore an important clinical need to prevent overtreatment and to enable earlier assignment of patients to more optimal therapies. Here we review some of the recent advances in the field by focusing on pathways mediating resistance to anti-HER2 therapies, and the role of the immune system and cancer stem cells in therapy response. We also review preoperative treatment strategies and research paradigms that show promise in identifying novel biomarkers of response while also enabling the delineation of the mechanisms underlying clinical benefit from anti-HER2 therapies.
- Published
- 2016
45. Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline Summary
- Author
-
Lyndsay N. Harris, Nofisat Ismaila, Lisa M. McShane, and Daniel F. Hayes
- Subjects
Oncology ,Oncology (nursing) ,Health Policy ,Clinical Decision-Making ,Humans ,Breast Neoplasms ,Female ,Neoplasm Invasiveness ,Combined Modality Therapy ,Biomarkers ,Neoplasm Staging - Published
- 2016
46. Brief-exposure to preoperative bevacizumab reveals a TGF-β signature predictive of response in HER2-negative breast cancers
- Author
-
Vinay, Varadan, Sitharthan, Kamalakaran, Hannah, Gilmore, Nilanjana, Banerjee, Angel, Janevski, Kristy L S, Miskimen, Nicole, Williams, Ajay, Basavanhalli, Anant, Madabhushi, Kimberly, Lezon-Geyda, Veerle, Bossuyt, Donald R, Lannin, Maysa, Abu-Khalaf, William, Sikov, Nevenka, Dimitrova, and Lyndsay N, Harris
- Subjects
Bevacizumab ,Receptor, ErbB-2 ,Sequence Analysis, RNA ,Transforming Growth Factor beta ,Humans ,Angiogenesis Inhibitors ,Breast Neoplasms ,Female ,Cell Hypoxia ,Signal Transduction - Abstract
To best define biomarkers of response, and to shed insight on mechanism of action of certain clinically important agents for early breast cancer, we used a brief-exposure paradigm in the preoperative setting to study transcriptional changes in patient tumors that occur with one dose of therapy prior to combination chemotherapy. Tumor biopsies from breast cancer patients enrolled in two preoperative clinical trials were obtained at baseline and after one dose of bevacizumab (HER2-negative), trastuzumab (HER2-positive) or nab-paclitaxel, followed by treatment with combination chemo-biologic therapy. RNA-Sequencing based PAM50 subtyping at baseline of 46 HER2-negative patients revealed a strong association between the basal-like subtype and pathologic complete response (pCR) to chemotherapy plus bevacizumab (p ≤ 0.0027), but did not provide sufficient specificity to predict response. However, a single dose of bevacizumab resulted in down-regulation of a well-characterized TGF-β activity signature in every single breast tumor that achieved pCR (p ≤ 0.004). The TGF-β signature was confirmed to be a tumor-specific read-out of the canonical TGF-β pathway using pSMAD2 (p ≤ 0.04), with predictive power unique to brief-exposure to bevacizumab (p ≤ 0.016), but not trastuzumab or nab-paclitaxel. Down-regulation of TGF-β activity was associated with reduction in tumor hypoxia by transcription and protein levels, suggesting therapy-induced disruption of an autocrine-loop between tumor stroma and malignant cells. Modulation of the TGF-β pathway upon brief-exposure to bevacizumab may provide an early functional readout of pCR to preoperative anti-angiogenic therapy in HER2-negative breast cancer, thus providing additional avenues for exploration in both preclinical and clinical settings with these agents.
- Published
- 2015
47. Phase 1b study of the mammalian target of rapamycin inhibitor sirolimus in combination with nanoparticle albumin-bound paclitaxel in patients with advanced solid tumors
- Author
-
Maysa M, Abu-Khalaf, Megan A, Baumgart, Scott N, Gettinger, Indukala, Doddamane, David P, Tuck, Shihe, Hou, Nianhang, Chen, Catherine, Sullivan, Kimberly, Lezon-Geyda, Daniel, Zelterman, Christos, Hatzis, Hari, Deshpande, Michael P, Digiovanna, Masoud, Azodi, Peter E, Schwartz, and Lyndsay N, Harris
- Subjects
Adult ,Male ,Sirolimus ,Paclitaxel ,TOR Serine-Threonine Kinases ,Middle Aged ,Cohort Studies ,Treatment Outcome ,Fluorodeoxyglucose F18 ,Albumins ,Neoplasms ,Positron-Emission Tomography ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Nanoparticles ,Female ,Albumin-Bound Paclitaxel ,Aged - Abstract
The optimal weekly oral dose of sirolimus and intravenous nanoparticle albumin-bound paclitaxel (nab-paclitaxel) were evaluated.A phase 1b study was performed to evaluate escalating doses of oral sirolimus (5-60 mg) on days 2, 9, and 16 with intravenous nab-paclitaxel (100 mg/m(2) ) on days 1, 8, and 15 in a 28-day cycle. A run-in treatment of nab-paclitaxel (day -14) and sirolimus (day -7) was administered for pharmacokinetic and pharmacodynamic assessments. Clinical trial endpoints included dose-limiting toxicities (DLTs), maximum tolerated doses, and response rates. Pharmacodynamics included immunohistochemistry for phosphatase and tensin homolog, mammalian target of rapamycin (mTOR), AKT, phosphorylated AKT, S6K1, and phosphorylated S6K1; exploratory gene expression analysis; and [(18) F]fludeoxyglucose (FDG) positron emission tomography.Twenty-three patients with advanced solid tumors were treated. Fifteen patients had prior taxane therapy. Twenty-two patients were evaluable for responses. One patient had a complete response, and 5 patients had a partial response (3 confirmed). DLTs were seen in 1 patient each at 10 (grade 3 dyspnea/hypoxia) and 40 mg (grade 4 leukopenia/neutropenia) and in 2 patients at 60 mg (grade 3 fatigue and grade 4 pericardial effusion). Patients with higher expression of posttreatment AKT and a greater decline in FDG activity were more likely to have a treatment response or stable disease.Sirolimus showed an acceptable safety profile at a weekly dose of 40 mg with weekly intravenous nab-paclitaxel at 100 mg/m(2) on days 1, 8, and 15 every 28 days. The posttreatment AKT score and changes in FDG activity may have roles as early predictors of responses to mTOR inhibitors.
- Published
- 2014
48. DTCs/CTCs in breast cancer: five decades later
- Author
-
Susan, Alsamarai, Maysa M, Abu-Khalaf, and Lyndsay N, Harris
- Subjects
Bone Marrow ,Neoplasm Micrometastasis ,Biomarkers, Tumor ,Humans ,Breast Neoplasms ,Female ,Neoplastic Cells, Circulating ,Prognosis ,Sensitivity and Specificity - Abstract
Since circulating tumor cells were first reported in 1955, the field has seen major advances in their detection and has established their prognostic impact. Here we review the current evidence for the prognostic and predictive value of circulating tumor cells in metastatic breast cancer. We then evaluate the role of CTCs and DTCs in early stage breast cancer. The weight of the evidence supports the role of CTCs and DTCs as prognostic indicators, however their role in therapy prediction remains unclear. Ongoing trials may provide answers and newer detection methods which improve sensitivity and specificity may have greater impact. At this point, the data does not support incorporation into clinical practice for early breast cancer patients.
- Published
- 2012
49. Circulating tumor cells in HER-2 positive metastatic breast cancer patients treated with trastuzumab and chemotherapy
- Author
-
Raquel A. Nunes, Xiaochun Li, Soonmo Peter Kang, Harold Burstein, Lisa Roberts, Walter Carney, Kimberly Blackwell, Paula Ryan, Virginia Borges, J. Dirk Iglehart, Paula Friedman, and Lyndsay N. Harris
- Subjects
0301 basic medicine ,Cancer Research ,Receptor, ErbB-2 ,Clinical Biochemistry ,Antineoplastic Agents ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Vinblastine ,Sensitivity and Specificity ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Piperidines ,Biomarkers, Tumor ,Humans ,DNA Primers ,Flavonoids ,Keratin-19 ,Base Sequence ,Immunomagnetic Separation ,Antibodies, Monoclonal ,Vinorelbine ,Trastuzumab ,Neoplastic Cells, Circulating ,Prognosis ,Protein Structure, Tertiary ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female - Abstract
The detection of circulating tumor cells (CTCs) in peripheral blood may have important prognostic and predictive implications in breast cancer treatment. A limitation in this field has been the lack of a validated method of accurately measuring CTCs. While sensitivity has improved using RT-PCR, specificity remains a major challenge. The goal of this paper is to present a sensitive and specific methodology of detecting CTCs in women with HER-2-positive metastatic breast cancer, and to examine its role as a marker that tracks disease response during treatment with trastuzumab-containing regimens. The study included patients with HER-2-positive metastatic breast cancer enrolled on two different clinical protocols using a trastuzumab-containing regimen. Serial CTCs were measured at planned time points and clinical correlations were made. Immunomagnetic selection of circulating epithelial cells was used to address the specificity of tumor cell detection using cytokeratin 19 (CK19). In addition, the extracellular domain of the HER-2 protein (HER-2/ECD) was measured to determine if CTCs detected by CK19 accurately reflect tumor burden. The presence of CTCs at first restaging was associated with disease progression. We observed an association between CK19 and HER-2/ECD. The association of HER-2/ECD with clinical response followed a similar pattern to that seen with CK19. Finally, the absence of HER-2/ECD at best overall response and a change of HER-2/ECD from positive at baseline to negative at best overall response was associated with favorable treatment response. Our study supports the prognostic and predictive role of the detection of CTCs in treatment of HER-2-positive metastatic breast cancer patients. The association between CK19 and markers of disease burden is in line with the concept that CTCs may be a reliable measure of tumor cells in the peripheral blood of patients with metastatic breast cancer. The association of CTCs at first restaging with treatment failure indicates that CTCs may have a role as surrogate markers to monitor treatment response.
- Published
- 2009
50. In Reply
- Author
-
Lyndsay N. Harris, Daniel F. Hayes, and Robert C. Bast
- Subjects
Cancer Research ,Oncology - Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.