24 results on '"Rimawi, M. F."'
Search Results
2. Endocrine‐based treatments in clinically‐relevant subgroups of hormone receptor‐positive/her2‐negative metastatic breast cancer: Systematic review and meta‐analysis
- Author
-
Schettini, F., Giuliano, M., Giudici, F., Conte, B., De Placido, P., Venturini, Sergio, Rognoni, C., Leo, A. D., Locci, M., Jerusalem, G., Mastro, L. D., Puglisi, F., Conte, P., De Laurentiis, M., Pusztai, L., Rimawi, M. F., Schiff, R., Arpino, G., De Placido, S., Prat, A., Generali, Daniele, Venturini S. (ORCID:0000-0002-6574-3337), Generali D. (ORCID:0000-0003-2480-3855), Schettini, F., Giuliano, M., Giudici, F., Conte, B., De Placido, P., Venturini, Sergio, Rognoni, C., Leo, A. D., Locci, M., Jerusalem, G., Mastro, L. D., Puglisi, F., Conte, P., De Laurentiis, M., Pusztai, L., Rimawi, M. F., Schiff, R., Arpino, G., De Placido, S., Prat, A., Generali, Daniele, Venturini S. (ORCID:0000-0002-6574-3337), and Generali D. (ORCID:0000-0003-2480-3855)
- Abstract
A precise assessment of the efficacy of first-/second-line endocrine therapies (ET) +/- target therapies (TT) in clinically-relevant subgroups of hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (MBC) has not yet been conducted. To improve our current knowledge and support clinical decision-making, we thus conducted a systematic literature search to identify all first-/second-line phase II/III randomized clinical trials (RCT) of currently approved or most promising ET +/- TT. Then, we performed a meta-analysis to assess progression-free (PFS) and/or overall survival (OS) benefit in several clinically-relevant prespecified subgroups. Thirty-five RCT were included (17,595 patients). Pooled results show significant reductions in the risk of relapse or death of 26-41% and 12-27%, respectively, depending on the clinical subgroup. Combination strategies proved to be more effective than single-agent ET (PFS hazard ratio (HR) range for combinations: 0.60-0.65 vs. HR range for single agent ET: 0.59-1.37; OS HR range for combinations: 0.74-0.87 vs. HR range for single agent ET: 0.68-0.98), with CDK4/6-inhibitors(i) + ET being the most effective regimen. Single agent ET showed comparable efficacy with ET+TT combinations in non-visceral (p = 0.63) and endocrine sensitive disease (p = 0.79), while mTORi-based combinations proved to be a valid therapeutic option in endocrine-resistant tumors, as well as PI3Ki + ET in PIK3CA-mutant tumors. These results strengthen international treatment guidelines and can aid therapeutic decision-making.
- Published
- 2021
3. Resistance to Anti-HER2 Therapies in Breast Cancer
- Author
-
Rimawi M. F., De Angelis C., Schiff R., Rimawi, M. F., De Angelis, C., and Schiff, R.
- Subjects
Antineoplastic Agent ,Receptors, Estrogen ,Animal ,Receptor, ErbB-2 ,Drug Resistance, Neoplasm ,Phosphatidylinositol 3-Kinase ,Breast Neoplasm ,Human - Abstract
HER2 is amplified or overexpressed in 20% to 25% of breast cancers. HER2 is a redundant, robust, and powerful signaling pathway that represents an attractive therapeutic target. Anti-HER2 therapy in the clinic has resulted in significant improvements in patient outcomes and, in recent years, combinations of anti-HER2 therapies have been explored and carry great promise. However, treatment resistance remains a problem. Resistance can be mediated, among others, by pathway redundancy, reactivation, or the utilization of escape pathways. Understanding mechanisms of resistance can lead to better therapeutic strategies to overcome resistance and optimize outcomes.
- Published
- 2015
4. Does incidence of rash predict treatment response to lapatanib in breast cancer?
- Author
-
Parma, J. M., primary, Pavlick, A. C., additional, Chang, J. C. N., additional, Rimawi, M. F., additional, and Trivedi, M., additional
- Published
- 2011
- Full Text
- View/download PDF
5. Triple-negative breast cancers: Biomarkers and outcomes.
- Author
-
Malorni, L., primary, Shetty, P. B., additional, Hilsenbeck, S. G., additional, Rimawi, M. F., additional, Elledge, R. M., additional, De Angelis, C., additional, Osborne, C. K., additional, De Placido, S., additional, and Arpino, G., additional
- Published
- 2010
- Full Text
- View/download PDF
6. EGFR expression in breast cancer: Association with biologic phenotype, prognosis, and resistance to adjuvant therapy
- Author
-
Rimawi, M. F., primary, Weiss, H. L., additional, Bhatia, P., additional, Chamness, G., additional, and Elledge, R. M., additional
- Published
- 2006
- Full Text
- View/download PDF
7. Activation of the IFN Signaling Pathway is Associated with Resistance to CDK4/6 Inhibitors and Immune Checkpoint Activation in ER-Positive Breast Cancer
- Author
-
Britta Weigelt, Resel Pereira, C. Kent Osborne, Rinath Jeselsohn, Jiangang Liu, Vidyalakshmi Sethunath, Jamunarani Veeraraghavan, Rachel Schiff, Lanfang Qin, Luca Malorni, Pier Selenica, Carmine De Angelis, Xiaoyong Fu, Ilenia Migliaccio, David N Brown, Susan G. Hilsenbeck, Sarmistha Nanda, Joshua Donaldson, Valerie M. Jansen, Sara A. Hurvitz, Agostina Nardone, Dennis J. Slamon, Ben Ho Park, Tao Wang, Jorge S. Reis-Filho, Lacey M. Litchfield, C. Guarducci, Matteo Benelli, Maria Letizia Cataldo, Mothaffar F. Rimawi, De Angelis, C., Fu, X., Cataldo, M. L., Nardone, A., Pereira, R., Veeraraghavan, J., Nanda, S., Qin, L., Sethunath, V., Wang, T., Hilsenbeck, S. G., Benelli, M., Migliaccio, I., Guarducci, C., Malorni, L., Litchfield, L. M., Liu, J., Donaldson, J., Selenica, P., Brown, D. N., Weigelt, B., Reis-Filho, J. S., Park, B. H., Hurvitz, S. A., Slamon, D. J., Rimawi, M. F., Jansen, V. M., Jeselsohn, R., Osborne, C. K., and Schiff, R.
- Subjects
0301 basic medicine ,Cancer Research ,Pyridines ,Oncology and Carcinogenesis ,Estrogen receptor ,Antineoplastic Agents ,Breast Neoplasms ,Palbociclib ,Piperazines ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Breast cancer ,Receptors ,Breast Cancer ,Genetics ,Tumor Cells, Cultured ,Humans ,Medicine ,Oncology & Carcinogenesis ,Cancer ,Cultured ,biology ,business.industry ,Kinase ,Cyclin-Dependent Kinase 4 ,Cyclin-Dependent Kinase 6 ,medicine.disease ,Estrogen ,Immune checkpoint ,Tumor Cells ,Good Health and Well Being ,030104 developmental biology ,Receptors, Estrogen ,Oncology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Cyclin-dependent kinase 6 ,Development of treatments and therapeutic interventions ,Signal transduction ,business ,Signal Transduction - Abstract
Purpose: Cyclin-dependent kinase 4 (CDK4) and CDK6 inhibitors (CDK4/6i) are highly effective against estrogen receptor–positive (ER+)/HER2− breast cancer; however, intrinsic and acquired resistance is common. Elucidating the molecular features of sensitivity and resistance to CDK4/6i may lead to identification of predictive biomarkers and novel therapeutic targets, paving the way toward improving patient outcomes. Experimental Design: Parental breast cancer cells and their endocrine-resistant derivatives (EndoR) were used. Derivatives with acquired resistance to palbociclib (PalboR) were generated from parental and estrogen deprivation–resistant MCF7 and T47D cells. Transcriptomic and proteomic analyses were performed in palbociclib-sensitive and PalboR lines. Gene expression data from CDK4/6i neoadjuvant trials and publicly available datasets were interrogated for correlations of gene signatures and patient outcomes. Results: Parental and EndoR breast cancer lines showed varying degrees of sensitivity to palbociclib. Transcriptomic analysis of these cell lines identified an association between high IFN signaling and reduced CDK4/6i sensitivity; thus an “IFN-related palbociclib-resistance Signature” (IRPS) was derived. In two neoadjuvant trials of CDK4/6i plus endocrine therapy, IRPS and other IFN-related signatures were highly enriched in patients with tumors exhibiting intrinsic resistance to CDK4/6i. PalboR derivatives displayed dramatic activation of IFN/STAT1 signaling compared with their short-term treated or untreated counterparts. In primary ER+/HER2− tumors, the IRPS score was significantly higher in lumB than lumA subtype and correlated with increased gene expression of immune checkpoints, endocrine resistance, and poor prognosis. Conclusions: Aberrant IFN signaling is associated with intrinsic resistance to CDK4/6i. Experimentally, acquired resistance to palbociclib is associated with activation of the IFN pathway, warranting additional studies to clarify its involvement in resistance to CDK4/6i.
- Published
- 2021
8. FOXA1 upregulation promotes enhancer and transcriptional reprogramming in endocrine-resistant breast cancer
- Author
-
Jorge S. Reis-Filho, Nikhil Wagle, Ofir Cohen, Sepideh Mehravaran, Jamunarani Veeraraghavan, Resel Pereira, Myles Brown, Lanfang Qin, Bert W. O'Malley, Carmine De Angelis, Carolina Gutierrez, Vidyalakshmi Sethunath, Xiaoyong Fu, Rachel Schiff, Maria Letizia Cataldo, Sarmistha Nanda, Qin Feng, Mothaffar F. Rimawi, Gary C. Chamness, Rinath Jeselsohn, Britta Weigelt, Pier Selenica, Agostina Nardone, C. Kent Osborne, Fu, X., Pereira, R., De Angelis, C., Veeraraghavan, J., Nanda, S., Qin, L., Cataldo, M. L., Sethunath, V., Mehravaran, S., Gutierrez, C., Chamness, G. C., Feng, Q., O'Malley, B. W., Selenica, P., Weigelt, B., Reis-Filho, J. S., Cohen, O., Wagle, N., Nardone, A., Jeselsohn, R., Brown, M., Rimawi, M. F., Osborne, C. K., and Schiff, R.
- Subjects
Multidisciplinary ,Chromatin binding ,Pioneer factor ,Enhancer/transcriptional reprogramming ,Biological Sciences ,Biology ,medicine.disease ,Metastatic breast cancer ,Metastasis ,Breast cancer ,Cancer research ,medicine ,FOXA1 ,Transcription factor ,Reprogramming ,Endocrine resistance - Abstract
Forkhead box A1 (FOXA1) is a pioneer factor that facilitates chromatin binding and function of lineage-specific and oncogenic transcription factors. Hyperactive FOXA1 signaling due to gene amplification or overexpression has been reported in estrogen receptor-positive (ER + ) endocrine-resistant metastatic breast cancer. However, the molecular mechanisms by which FOXA1 up-regulation promotes these processes and the key downstream targets of the FOXA1 oncogenic network remain elusive. Here, we demonstrate that FOXA1 overexpression in ER + breast cancer cells drives genome-wide enhancer reprogramming to activate prometastatic transcriptional programs. Up-regulated FOXA1 employs superenhancers (SEs) to synchronize transcriptional reprogramming in endocrine-resistant breast cancer cells, reflecting an early embryonic development process. We identify the hypoxia-inducible transcription factor hypoxia-inducible factor-2α (HIF-2α) as the top high FOXA1-induced SE target, mediating the impact of high FOXA1 in activating prometastatic gene sets and pathways associated with poor clinical outcome. Using clinical ER + /HER2 − metastatic breast cancer datasets, we show that the aberrant FOXA1/HIF-2α transcriptional axis is largely nonconcurrent with the ESR1 mutations, suggesting different mechanisms of endocrine resistance and treatment strategies. We further demonstrate the selective efficacy of an HIF-2α antagonist, currently in clinical trials for advanced kidney cancer and recurrent glioblastoma, in reducing the clonogenicity, migration, and invasion of endocrine-resistant breast cancer cells expressing high FOXA1. Our study has uncovered high FOXA1-induced enhancer reprogramming and HIF-2α–dependent transcriptional programs as vulnerable targets for treating endocrine-resistant and metastatic breast cancer.
- Published
- 2019
9. A combinatorial biomarker predicts pathologic complete response to neoadjuvant lapatinib and trastuzumab without chemotherapy in patients with HER2+ breast cancer
- Author
-
Rachel Schiff, Mothaffar F. Rimawi, Matthew P. Goetz, Jorge S. Reis-Filho, Ting Wang, Britta Weigelt, Carolina Gutierrez, CK Osborne, Aleix Prat, Suzanne A. W. Fuqua, Antonio C. Wolff, R. Mao, Jamunarani Veeraraghavan, Andres Forero, Anna C. Pavlick, Paolo Nuciforo, Ingrid A. Mayer, C. De Angelis, SG Hilsenbeck, Jenny C. Chang, Ian E. Krop, Rita Nanda, Sabrina Herrera, Alejandro Contreras, De Angelis, C., Veeraraghavan, J., Mao, R., Wang, T., Herrera, S., Pavlick, A. C., Contreras, A., Nuciforo, P., Mayer, I. A., Forero, A., Nanda, R., Goetz, M. P., Chang, J. C., Wolff, A. C., Krop, I. E., Fuqua, S. A. W., Prat, A., Hilsenbeck, S. G., Weigelt, B., Reis-Filho, J. S., Gutierrez, C., Osborne, C. K., Rimawi, M. F., and Schiff, R.
- Subjects
0301 basic medicine ,Oncology ,Receptor, ErbB-2 ,medicine.medical_treatment ,Estrogen receptor ,PIK3CA mutation ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,PTEN protein ,skin and connective tissue diseases ,In Situ Hybridization, Fluorescence ,Neoadjuvant therapy ,biology ,Remission Induction ,Hematology ,Prognosis ,Chemotherapy regimen ,Neoadjuvant Therapy ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,precision medicine ,Breast Neoplasms ,Lapatinib ,03 medical and health sciences ,breast cancer ,Breast cancer ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,PTEN ,neoplasms ,Chemotherapy ,business.industry ,ErbB2 receptor tyrosine kinase ,Gene Amplification ,Original Articles ,medicine.disease ,030104 developmental biology ,fluorescent in situ hybridization ,biology.protein ,business ,Follow-Up Studies - Abstract
BACKGROUND: HER2-positive (+) breast cancers, defined by HER2 overexpression and/or amplification, are often addicted to HER2 to maintain their malignant phenotype. Yet, some HER2+ tumors do not benefit from anti-HER2 therapy. We hypothesize that HER2 amplification levels and PI3K pathway activation are key determinants of response to HER2-targeted treatments without chemotherapy. PATIENTS AND METHODS: Baseline HER2+ tumors from patients treated with neoadjuvant lapatinib plus trastuzumab [with endocrine therapy for estrogen receptor (ER)+ tumors] in TBCRC006 (NCT00548184) were evaluated in a central laboratory for HER2 amplification by fluorescence in situ hybridization (FISH) (n = 56). HER2 copy number (CN) and FISH ratios, and PI3K pathway status, defined by PIK3CA mutations or PTEN levels by immunohistochemistry were available for 41 tumors. Results were correlated with pathologic complete response (pCR; no residual invasive tumor in breast). RESULTS: Thirteen of the 56 patients (23%) achieved pCR. None of the 11 patients with HER2 ratio
- Published
- 2019
10. The oral selective oestrogen receptor degrader (SERD) AZD9496 is comparable to fulvestrant in antagonising ER and circumventing endocrine resistance
- Author
-
Agostina Nardone, Rachel Schiff, Henry Brown, Meghana V. Trivedi, Martin Shea, Mark Pilling, Jamunarani Veeraraghavan, Chandandeep Nagi, Rinath Jeselsohn, Carmine De Angelis, Oona Delpuech, Maria Letizia Cataldo, Tamika Mitchell, C. Kent Osborne, Mothaffar F. Rimawi, Hazel M. Weir, Gary C. Chamness, Xiaoyong Fu, Susan G. Hilsenbeck, Pilling, Mark [0000-0002-7446-6597], Apollo - University of Cambridge Repository, Nardone, A., Weir, H., Delpuech, O., Brown, H., De Angelis, C., Cataldo, M. L., Fu, X., Shea, M. J., Mitchell, T., Veeraraghavan, J., Nagi, C., Pilling, M., Rimawi, M. F., Trivedi, M., Hilsenbeck, S. G., Chamness, G. C., Jeselsohn, R., Osborne, C. K., and Schiff, R.
- Subjects
Cancer Research ,Indoles ,Neoplasms, Hormone-Dependent ,Breast Neoplasms ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,MCF-7 Cell ,0302 clinical medicine ,Breast cancer ,Cinnamate ,In vivo ,medicine ,Animals ,Humans ,Receptor ,skin and connective tissue diseases ,Fulvestrant ,Cancer ,Cell Proliferation ,Estradiol ,Animal ,Cell growth ,Estrogens ,medicine.disease ,Estrogen ,Metastatic breast cancer ,3. Good health ,Tamoxifen ,Oncology ,chemistry ,Receptors, Estrogen ,Indole ,Cell culture ,Cinnamates ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,MCF-7 Cells ,Heterografts ,Female ,Growth inhibition ,Heterograft ,Breast Neoplasm ,Human ,medicine.drug - Abstract
BACKGROUND: The oestrogen receptor (ER) is an important therapeutic target in ER-positive (ER+) breast cancer. The selective ER degrader (SERD), fulvestrant, is effective in patients with metastatic breast cancer, but its intramuscular route of administration and low bioavailability are major clinical limitations. METHODS: Here, we studied the pharmacology of a new oral SERD, AZD9496, in a panel of in vitro and in vivo endocrine-sensitive and -resistant breast cancer models. RESULTS: In endocrine-sensitive models, AZD9496 inhibited cell growth and blocked ER activity in the presence or absence of oestrogen. In vivo, in the presence of oestrogen, short-term AZD9496 treatment, like fulvestrant, resulted in tumour growth inhibition and reduced expression of ER-dependent genes. AZD9496 inhibited cell growth in oestrogen deprivation-resistant and tamoxifen-resistant cell lines and xenograft models that retain ER expression. AZD9496 effectively reduced ER levels and ER-induced transcription. Expression analysis of short-term treated tumours showed that AZD9496 potently inhibited classic oestrogen-induced gene transcription, while simultaneously increasing expression of genes negatively regulated by ER, including genes potentially involved in escape pathways of endocrine resistance. CONCLUSIONS: These data suggest that AZD9496 is a potent anti-oestrogen that antagonises and degrades ER with anti-tumour activity in both endocrine-sensitive and endocrine-resistant models.
- Published
- 2018
11. Neratinib plus trastuzumab is superior to pertuzumab plus trastuzumab in HER2-positive breast cancer xenograft models
- Author
-
Alshad S. Lalani, Sarmistha Nanda, Mario Giuliano, Tamika Mitchell, Jamunarani Veeraraghavan, Carmine De Angelis, C. Kent Osborne, Martin Shea, Chandandeep Nagi, Sepideh Mehravaran, Lanfang Qin, Kristina Goutsouliak, Irmina Diala, Robert Davis, Mothaffar F. Rimawi, Tao Wang, Susan G. Hilsenbeck, Vidyalakshmi Sethunath, Rachel Schiff, Resel Pereira, Carolina Gutierrez, Veeraraghavan, J., Gutierrez, C., Sethunath, V., Mehravaran, S., Giuliano, M., Shea, M. J., Mitchell, T., Wang, T., Nanda, S., Pereira, R., Davis, R., Goutsouliak, K., Qin, L., De Angelis, C., Diala, I., Lalani, A. S., Nagi, C., Hilsenbeck, S. G., Rimawi, M. F., Osborne, C. K., and Schiff, R.
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Estrogen receptor ,Lapatinib ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Targeted therapies ,Trastuzumab ,medicine ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,skin and connective tissue diseases ,RC254-282 ,Chemotherapy ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Neratinib ,Cancer research ,Immunohistochemistry ,Pertuzumab ,business ,medicine.drug - Abstract
Lapatinib (L) plus trastuzumab (T), with endocrine therapy for estrogen receptor (ER)+ tumors, but without chemotherapy, yielded meaningful response in HER2+ breast cancer (BC) neoadjuvant trials. The irreversible/pan-HER inhibitor neratinib (N) has proven more potent than L. However, the efficacy of N+T in comparison to pertuzumab (P) + T or L + T (without chemotherapy) remains less studied. To address this, mice bearing HER2+ BT474-AZ (ER+) cell and BCM-3963 patient-derived BC xenografts were randomized to vehicle, N, T, P, N+T, or P+T, with simultaneous estrogen deprivation for BT474-AZ. Time to tumor regression/progression and incidence/time to complete response (CR) were determined. Changes in key HER pathway and proliferative markers were assessed by immunohistochemistry and western blot of short-term-treated tumors. In the BT474-AZ model, while all N, P, T, N + T, and P + T treated tumors regressed, N + T-treated tumors regressed faster than P, T, and P + T. Further, N + T was superior to N and T alone in accelerating CR. In the BCM-3963 model, which was refractory to T, P, and P + T, while N and N + T yielded 100% CR, N + T accelerated the CR compared to N. Ki67, phosphorylated (p) AKT, pS6, and pERK levels were largely inhibited by N and N + T, but not by T, P, or P + T. Phosphorylated HER receptor levels were also markedly inhibited by N and N + T, but not by P + T or L + T. Our findings establish the efficacy of combining N with T and support clinical testing to investigate the efficacy of N + T with or without chemotherapy in the neoadjuvant setting for HER2+ BC.
- Published
- 2021
12. Endocrine‐based treatments in clinically‐relevant subgroups of hormone receptor‐positive/her2‐negative metastatic breast cancer: Systematic review and meta‐analysis
- Author
-
Mothaffar F. Rimawi, Grazia Arpino, Angelo Di Leo, Fabiola Giudici, Pietro De Placido, Sergio Venturini, Mario Giuliano, Lajos Pusztai, Sabino De Placido, Lucia Del Mastro, Guy Jerusalem, Daniele Generali, Michelino De Laurentiis, Fabio Puglisi, Francesco Schettini, Carla Rognoni, Aleix Prat, Benedetta Conte, Rachel Schiff, Mariavittoria Locci, Pierfranco Conte, Schettini, F., Giuliano, M., Giudici, F., Conte, B., De Placido, P., Venturini, S., Rognoni, C., Leo, A. D., Locci, M., Jerusalem, G., Mastro, L. D., Puglisi, F., Conte, P., De Laurentiis, M., Pusztai, L., Rimawi, M. F., Schiff, R., Arpino, G., De Placido, S., Prat, A., Generali, D., Schettini, Francesco, Giuliano, Mario, Giudici, Fabiola, Conte, Benedetta, De Placido, Pietro, Venturini, Sergio, Rognoni, Carla, Di Leo, Angelo, Locci, Mariavittoria, Jerusalem, Guy, Del Mastro, Lucia, Puglisi, Fabio, Conte, Pierfranco, De Laurentiis, Michelino, Pusztai, Lajo, Rimawi, Mothaffar F., Schiff, Rachel, Arpino, Grazia, De Placido, Sabino, Prat, Aleix, and Generali, Daniele
- Subjects
Oncology ,Endocrine therapy ,Cancer Research ,medicine.medical_specialty ,Disease ,lcsh:RC254-282 ,meta-analysi ,Meta‐analysi ,Hormone receptor ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,systematic review ,law ,Internal medicine ,medicine ,Endocrine system ,030212 general & internal medicine ,ENDOCRINE THERAPY, HORMONE RECEPTOR, METASTATIC BREAST CANCER, META‐ANALYSIS, SYSTEMATIC REVIEW ,business.industry ,endocrine therapy ,Hazard ratio ,hormone receptor ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Metastatic breast cancer ,Meta‐analysis ,Systematic review ,meta-analysis ,Regimen ,Settore SECS-S/01 - STATISTICA ,030220 oncology & carcinogenesis ,Meta-analysis ,metastatic breast cancer ,business ,Hormone - Abstract
Simple Summary Hormone receptor-positive (HR+)/HER2-negative is the most frequent subgroup of metastatic breast cancer (MBC). Important therapeutic advances in the treatment of this tumor type have been observed in the last 20 years, with the approval of numerous endocrine therapies (ET) with or without target therapies (TT). To improve our current knowledge and support clinical decision-making, we conducted a systematic literature and meta-analysis focused on the most relevant/promising first-/second-line ET ± TT of the last 20 years. We observed that CDK4/6-inhibitors(i) + ET were the most effective regimens. At the same time, mTORi-based combinations proved to be a valid therapeutic option in endocrine-resistant tumors, as well as PI3Ki + ET in PIK3CA-mutant patients. Single agent ET might still be a valuable upfront treatment in endocrine sensitive and non-visceral disease. Abstract A precise assessment of the efficacy of first-/second-line endocrine therapies (ET) ± target therapies (TT) in clinically-relevant subgroups of hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (MBC) has not yet been conducted. To improve our current knowledge and support clinical decision-making, we thus conducted a systematic literature search to identify all first-/second-line phase II/III randomized clinical trials (RCT) of currently approved or most promising ET ± TT. Then, we performed a meta-analysis to assess progression-free (PFS) and/or overall survival (OS) benefit in several clinically-relevant prespecified subgroups. Thirty-five RCT were included (17,595 patients). Pooled results show significant reductions in the risk of relapse or death of 26–41% and 12–27%, respectively, depending on the clinical subgroup. Combination strategies proved to be more effective than single-agent ET (PFS hazard ratio (HR) range for combinations: 0.60–0.65 vs. HR range for single agent ET: 0.59–1.37; OS HR range for combinations: 0.74–0.87 vs. HR range for single agent ET: 0.68–0.98), with CDK4/6-inhibitors(i) + ET being the most effective regimen. Single agent ET showed comparable efficacy with ET+TT combinations in non-visceral (p = 0.63) and endocrine sensitive disease (p = 0.79), while mTORi-based combinations proved to be a valid therapeutic option in endocrine-resistant tumors, as well as PI3Ki + ET in PIK3CA-mutant tumors. These results strengthen international treatment guidelines and can aid therapeutic decision-making.
- Published
- 2021
13. HER2-enriched subtype and pathological complete response in HER2-positive breast cancer: a systematic review and meta-analysis
- Author
-
Aranzazu Fernandez-Martinez, Barbara Adamo, Blanca Gonzalez-Farre, Lisa A. Carey, Francesco Schettini, Benedetta Conte, Charles M. Perou, Jamunarani Veeraraghavan, Jan C. Brase, Sabino De Placido, Tomás Pascual, Mariavittoria Locci, Pierfranco Conte, Patricia Villagrasa, Montserrat Muñoz, Sonia Pernas, Fara Brasó-Maristany, Olga Martínez, Patricia Galván, Maria Vidal, Mothaffar F. Rimawi, C. Kent Osborne, Carla Rognoni, Gaia Griguolo, Rachel Schiff, Nuria Chic, Valentina Guarneri, Aleix Prat, Schettini, F., Pascual, T., Conte, B., Chic, N., Braso-Maristany, F., Galvan, P., Martinez, O., Adamo, B., Vidal, M., Munoz, M., Fernandez-Martinez, A., Rognoni, C., Griguolo, G., Guarneri, V., Conte, P. F., Locci, M., Brase, J. C., Gonzalez-Farre, B., Villagrasa, P., De Placido, S., Schiff, R., Veeraraghavan, J., Rimawi, M. F., Osborne, C. K., Pernas, S., Perou, C. M., Carey, L. A., and Prat, A.
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,BIOMARKER, BREAST CANCER, HER2-ENRICHED, HER2-POSITIVE, PAM50, PATHOLOGIC COMPLETE RESPONSE ,Receptor, ErbB-2 ,medicine.medical_treatment ,Breast Neoplasms ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,ErbB-2 ,Internal medicine ,Pathologic complete response ,medicine ,Biomarkers, Tumor ,Humans ,Radiology, Nuclear Medicine and imaging ,PAM50 ,skin and connective tissue diseases ,neoplasms ,Neoadjuvant therapy ,Neoplasm Staging ,Chemotherapy ,Tumor ,business.industry ,Remission Induction ,Biomarker ,HER2-Enriched ,HER2-positive ,Female ,Neoadjuvant Therapy ,General Medicine ,Odds ratio ,medicine.disease ,Genomic Biomarker ,030104 developmental biology ,030220 oncology & carcinogenesis ,Meta-analysis ,Biomarker (medicine) ,business ,Biomarkers ,Receptor - Abstract
Background: HER2-positive (HER2+) breast cancer (BC) comprises all the four PAM50 molecular subtypes. Among these, the HER2-Enriched (HER2-E) appear to be associated with higher pathological complete response (pCR) rates following anti-HER2-based regimens. Here, we present a meta-analysis to validate the association of the HER2-E subtype with pCR following anti-HER2-based neoadjuvant treatments with or without chemotherapy (CT). Methods: A systematic literature search was performed in February 2019. The primary objective was to compare the association between HER2-E subtype (versus others) and pCR. Selected secondary objectives were to compare the association between 1) HER2-E subtype and pCR in CT-free studies, 2) HER2-E subtype within hormone receptor (HR)-negative and HR+ disease and 3) HR-negative disease (versus HR+) and pCR in all patients and within HER2-E subtype. A random-effect model was applied. The Higgins��� I2 was used to quantify heterogeneity. Results: Sixteen studies were included, 5 of which tested CT-free regimens. HER2-E subtype was significantly associated with pCR in all patients (odds ratio [OR] = 3.50, p < 0.001, I2 = 33%), in HR+ (OR = 3.61, p < 0.001, I2 = 1%) and HR-negative tumors (OR = 2.28, p = 0.01, I2 = 47%). In CT-free studies, HER2-E subtype was associated with pCR in all patients (OR = 5.52, p < 0.001, I2 = 0%) and in HR + disease (OR = 4.08, p = 0.001, I2 = 0%). HR-negative status was significantly associated with pCR compared to HR + status in all patients (OR = 2.41, p < 0.001, I2 = 30%) and within the HER2-E subtype (OR = 1.76, p < 0.001, I2 = 0%). Conclusions: The HER2-E biomarker identifies patients with a higher likelihood of achieving a pCR following neoadjuvant anti-HER2-based therapy beyond HR status and CT use. Future trial designs to escalate or de-escalate systemic therapy in HER2+ disease should consider this genomic biomarker.
- Published
- 2020
14. Towards personalized treatment for early stage HER2-positive breast cancer
- Author
-
C. Kent Osborne, Vidyalakshmi Sethunath, Rachel Schiff, Mothaffar F. Rimawi, Jamunarani Veeraraghavan, Carmine De Angelis, Kristina Goutsouliak, Goutsouliak, K., Veeraraghavan, J., Sethunath, V., De Angelis, C., Osborne, C. K., Rimawi, M. F., and Schiff, R.
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Receptor, ErbB-2 ,medicine.medical_treatment ,Breast Neoplasms ,Disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Adjuvant therapy ,Humans ,Molecular Targeted Therapy ,Precision Medicine ,Intensive care medicine ,skin and connective tissue diseases ,neoplasms ,Neoplasm Staging ,Chemotherapy ,Antineoplastic Combined Chemotherapy Protocol ,business.industry ,Precision medicine ,medicine.disease ,Clinical trial ,030104 developmental biology ,Oncology ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,Localized disease ,Female ,business ,Breast Neoplasm ,medicine.drug ,Human - Abstract
Advances in HER2-targeted therapies have improved the survival of patients with HER2-positive breast cancer. The standard-of-care treatment for localized disease has been chemotherapy and 1 year of adjuvant HER2-targeted therapy, typically with the anti-HER2 antibody trastuzumab. Despite the effectiveness of this treatment, disease relapse occurs in a subset of patients; thus, focus has been placed on escalating treatment by either combining different HER2-targeted agents or extending the duration of HER2-targeted therapy. Indeed, dual HER2-targeted therapies and extended-duration anti-HER2 therapy, as well as adjuvant therapy with the anti-HER2 antibody–drug conjugate T-DM1, have all been approved for clinical use. Emerging evidence suggests, however, that some patients do not derive sufficient benefit from these additional therapies to offset the associated toxicities and/or costs. Similarly, the universal use of chemotherapy might not benefit all patients, and treatment de-escalation through omission of chemotherapy has shown promise in clinical trials and is currently being explored further. The future of precision medicine should therefore involve tailoring of therapy based on the genetics and biology of each tumour and the clinical characteristics of each patient. Predictive biomarkers that enable the identification of patients who will benefit from either escalated or de-escalated treatment will be crucial to this approach. In this Review, we summarize the available HER2-targeted agents and associated mechanisms of resistance, and describe the current therapeutic landscape of early stage HER2-positive breast cancer, focusing on strategies for treatment escalation or de-escalation. HER2-targeted therapy has greatly improved the outcomes of patients with HER2-positive breast cancer, with a range of agents now approved or in late-stage clinical development. In the era of precision medicine, efforts are being made to further improve patient outcomes by personalizing HER2-targeted treatment regimens, primarily though escalation or de-escalation of therapy according to the disease biology. In this Review, the authors provide an overview of the current landscape of HER2-targeted therapy and discuss the evidence supporting such tailored therapeutic strategies.
- Published
- 2020
15. Low PTEN levels and PIK3CA mutations predict resistance to neoadjuvant lapatinib and trastuzumab without chemotherapy in patients with HER2 over-expressing breast cancer
- Author
-
Britta Weigelt, Anne Pavlick, C. Kent Osborne, Marilyn M. Li, Kathleen A. Burke, Jorge S. Reis-Filho, Tao Wang, Fresia Pareja, Matthew P. Goetz, Rachel Schiff, Jenny C. Chang, Rita Nanda, Suzanne A. W. Fuqua, Carmine De Angelis, Ian E. Krop, Ingrid A. Mayer, Mothaffar F. Rimawi, Sabrina Herrera, Antonio C. Wolff, Andres Forero, Felipe C Geyer, Alejandro Contreras, Susan G. Hilsenbeck, Carolina Gutierrez, Rimawi, M. F., de Angelis, C., Contreras, A., Pareja, F., Geyer, F. C., Burke, K. A., Herrera, S., Wang, T., Mayer, I. A., Forero, A., Nanda, R., Goetz, M. P., Chang, J. C., Krop, I. E., Wolff, A. C., Pavlick, A. C., Fuqua, S. A. W., Gutierrez, C., Hilsenbeck, S. G., Li, M. M., Weigelt, B., Reis-Filho, J. S., Osborne, C. K., and Schiff, R.
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Receptor, ErbB-2 ,medicine.medical_treatment ,Estrogen receptor ,PIK3CA mutation ,0302 clinical medicine ,Trastuzumab ,Antineoplastic Combined Chemotherapy Protocols ,skin and connective tissue diseases ,Class I Phosphatidylinositol 3-Kinase ,biology ,Middle Aged ,Neoadjuvant Therapy ,Gene Expression Regulation, Neoplastic ,PCR ,030220 oncology & carcinogenesis ,Immunohistochemistry ,Female ,Breast Neoplasm ,Human ,medicine.drug ,Adult ,medicine.medical_specialty ,Class I Phosphatidylinositol 3-Kinases ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Lapatinib ,Article ,03 medical and health sciences ,Breast cancer ,Internal medicine ,medicine ,Humans ,PTEN ,HER2-positive breast cancer ,neoplasms ,PI3K/AKT/mTOR pathway ,Aged ,Chemotherapy ,Antineoplastic Combined Chemotherapy Protocol ,business.industry ,PTEN Phosphohydrolase ,Quinazoline ,medicine.disease ,PTEN level ,030104 developmental biology ,Mutation ,Quinazolines ,biology.protein ,business - Abstract
Aberrant activation of the PI3K pathway has been implicated in resistance to HER2-targeted therapy, but results of clinical trials are confounded by the co-administration of chemotherapy. We investigated the effect of perturbations of this pathway in breast cancers from patients treated with neoadjuvant anti-HER2-targeted therapy without chemotherapy. Baseline tumor samples from patients with HER2-positive breast cancer enrolled in TBCRC006 (NCT00548184), a 12-week neoadjuvant clinical trial with lapatinib plus trastuzumab [plus endocrine therapy for estrogen receptor (ER)-positive tumors], were assessed for PTEN status by immunohistochemistry and PIK3CA mutations by sequencing. Results were correlated with pathologic complete response (pCR). Of 64 evaluable patients, PTEN immunohistochemistry and PIK3CA mutation analysis were performed for 59 and 46 patients, respectively. PTEN status (dichotomized by H-score median) was correlated with pCR (32% in high PTEN vs. 9% in low PTEN, p = 0.04). PIK3CA mutations were identified in 14/46 tumors at baseline (30%) and did not correlate with ER or PTEN status. One patient whose tumor harbored a PIK3CA mutation achieved pCR (p = 0.14). When considered together (43 cases), 1/25 cases (4%) with a PIK3CA mutation and/or low PTEN expression levels had a pCR compared to 7/18 cases (39%) with wild-type PI3KCA and high PTEN expression levels (p = 0.006). PI3K pathway activation is associated with resistance to lapatinib and trastuzumab in breast cancers, without chemotherapy. Further studies are warranted to investigate how to use these biomarkers to identify upfront patients who may respond to anti-HER2 alone, without chemotherapy.
- Published
- 2017
16. De-escalation of treatment in HER2-positive breast cancer: Determinants of response and mechanisms of resistance
- Author
-
C. Kent Osborne, Carmine De Angelis, Jorge S. Reis-Filho, Jamunarani Veeraraghavan, Aleix Prat, Mothaffar F. Rimawi, Rachel Schiff, Tomás Pascual, Veeraraghavan, J., De Angelis, C., Reis-Filho, J. S., Pascual, T., Prat, A., Rimawi, M. F., Osborne, C. K., and Schiff, R.
- Subjects
0301 basic medicine ,PI3K/PTEN ,Receptor, ErbB-2 ,Estrogen receptor ,HER2-targeted therapy resistance ,Antineoplastic Agents ,Breast Neoplasms ,Disease ,Article ,Metastasis ,Antineoplastic Agent ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Trastuzumab ,medicine ,Oncogene Addiction ,Tumor Microenvironment ,Humans ,HER2-positive breast cancer ,skin and connective tissue diseases ,PI3K/AKT/mTOR pathway ,Tumor microenvironment ,business.industry ,Oncogenic addiction ,Quinazoline ,Lapatinib ,General Medicine ,medicine.disease ,Clinical trial ,030104 developmental biology ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Quinazolines ,Surgery ,Female ,Signal transduction ,Phosphatidylinositol 3-Kinase ,business ,Proto-Oncogene Proteins c-akt ,Breast Neoplasm ,medicine.drug ,Human ,Signal Transduction - Abstract
Overexpression and/or gene amplification of HER2, a crucial member of the HER family of four receptors, occur in about 15-20% of breast cancers and define an aggressive subtype of the disease. Activated HER homo and heterodimers govern a complex and redundant downstream signaling network that regulates cell survival and metastasis. Despite treatment with effective HER2-targeted therapies, many HER2-positive tumors fail to respond, or initially respond but eventually develop resistance. One of the upfront reasons for this treatment failure is failure to accurately select the tumors that are truly dependent on HER2 for survival and so would benefit the most from HER2-targeted therapy. In these truly HER2-addicted tumors (i.e. physiologically dependent), resistance could be the result of an incomplete inhibition of signaling at the HER receptor layer. In this regard, preclinical and clinical studies have documented the superiority of combination anti-HER2 therapy over single agent therapy to achieve a more comprehensive inhibition of the various HER receptor dimers. HER2 can be further activated or reactivated by mutations or other alterations in HER2 itself, or in other HER family members. Even when a complete and sustained HER inhibition is achieved, resistance to anti-HER therapy can arise by other somewhat dominant mechanisms, including preexisting or emerging alternative signaling pathways such as the estrogen receptor, deregulated downstream signaling components, especially of the PI3K pathway, and the tumor immune microenvironment. Most of the clinical trials that have investigated the efficacy of anti-HER2 therapies took place in the background of aggressive chemotherapy regimens, thus confounding the identification of key factors of resistance to the anti-HER2 treatments. Recent studies, however, have suggested that some HER2-amplified tumors may benefit from anti-HER2 therapy combined with only a single chemotherapy agent or in the absence of any chemotherapy. This de-escalation approach, a promising therapeutic strategy, is currently being explored in the clinic. In this review, we summarize the major molecular determinants that play a crucial role in influencing tumor response and resistance to HER2-targeted therapy, and discuss the growing need for patient stratification in order to facilitate the development of de-escalation strategies using HER2-targeted therapy alone with no chemotherapy.
- Published
- 2017
17. HER2 Reactivation through Acquisition of the HER2 L755S Mutation as a Mechanism of Acquired Resistance to HER2-targeted Therapy in HER2+ Breast Cancer
- Author
-
Kathleen A. Burke, Ian Waters, Agostina Nardone, Joe W. Gray, Martin Shea, Ben Ho Park, Carmine De Angelis, Sarmistha Nanda, Chandandeep Nagi, Daniel J. Zabransky, Mahitha Rajendran, Jamunarani Veeraraghavan, Felipe C Geyer, Tamika Mitchell, Carolina Gutierrez, Huizhong Hu, Gary C. Chamness, Britta Weigelt, C. Kent Osborne, Jorge S. Reis-Filho, Nicholas J. Wang, Vidyalakshmi Sethunath, Kenneth L. Scott, Rachel Schiff, Lanfang Qin, Xiaowei Xu, Alexander Renwick, Susan G. Hilsenbeck, Laura M. Heiser, Mothaffar F. Rimawi, Edward S. Chen, Chad A. Shaw, Charlotte K.Y. Ng, Tao Wang, Xu, X., De Angelis, C., Burke, K. A., Nardone, A., Hu, H., Qin, L., Veeraraghavan, J., Sethunath, V., Heiser, L. M., Wang, N., Ng, C. K. Y., Chen, E. S., Renwick, A., Wang, T., Nanda, S., Shea, M., Mitchell, T., Rajendran, M., Waters, I., Zabransky, D. J., Scott, K. L., Gutierrez, C., Nagi, C., Geyer, F. C., Chamness, G. C., Park, B. H., Shaw, C. A., Hilsenbeck, S. G., Rimawi, M. F., Gray, J. W., Weigelt, B., Reis-Filho, J. S., Osborne, C. K., and Schiff, R.
- Subjects
0301 basic medicine ,Cancer Research ,Xenograft Model Antitumor Assay ,Receptor, ErbB-2 ,Afatinib ,medicine.medical_treatment ,Breast Neoplasms ,Biology ,Lapatinib ,Antibodies, Monoclonal, Humanized ,Article ,Targeted therapy ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Germline mutation ,Breast cancer ,Trastuzumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,skin and connective tissue diseases ,Cell Proliferation ,Animal ,Cancer ,Quinazoline ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,Receptors, Estrogen ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Immunology ,Neratinib ,Mutation ,Cancer research ,Quinazolines ,Female ,Breast Neoplasm ,medicine.drug ,Human ,Signal Transduction - Abstract
Purpose: Resistance to anti-HER2 therapies in HER2+ breast cancer can occur through activation of alternative survival pathways or reactivation of the HER signaling network. Here we employed BT474 parental and treatment-resistant cell line models to investigate a mechanism by which HER2+ breast cancer can reactivate the HER network under potent HER2-targeted therapies.Experimental Design: Resistant derivatives to lapatinib (L), trastuzumab (T), or the combination (LR/TR/LTR) were developed independently from two independent estrogen receptor ER+/HER2+ BT474 cell lines (AZ/ATCC). Two derivatives resistant to the lapatinib-containing regimens (BT474/AZ-LR and BT474/ATCC-LTR lines) that showed HER2 reactivation at the time of resistance were subjected to massive parallel sequencing and compared with parental lines. Ectopic expression and mutant-specific siRNA interference were applied to analyze the mutation functionally. In vitro and in vivo experiments were performed to test alternative therapies for mutant HER2 inhibition.Results: Genomic analyses revealed that the HER2L755S mutation was the only common somatic mutation gained in the BT474/AZ-LR and BT474/ATCC-LTR lines. Ectopic expression of HER2L755S induced acquired lapatinib resistance in the BT474/AZ, SK-BR-3, and AU565 parental cell lines. HER2L755S-specific siRNA knockdown reversed the resistance in BT474/AZ-LR and BT474/ATCC-LTR lines. The HER1/2–irreversible inhibitors afatinib and neratinib substantially inhibited both resistant cell growth and the HER2 and downstream AKT/MAPK signaling driven by HER2L755S in vitro and in vivo.Conclusions: HER2 reactivation through acquisition of the HER2L755S mutation was identified as a mechanism of acquired resistance to lapatinib-containing HER2-targeted therapy in preclinical HER2-amplified breast cancer models, which can be overcome by irreversible HER1/2 inhibitors. Clin Cancer Res; 23(17); 5123–34. ©2017 AACR.
- Published
- 2017
18. FOXA1 overexpression mediates endocrine resistance by altering the ER transcriptome and IL-8 expression in ER-positive breast cancer
- Author
-
Xiaoyong Fu, Myles Brown, Rachel Schiff, Carolina Gutierrez, Laura M. Heiser, Martin Shea, C. Kent Osborne, Obi L. Griffith, Anna Tsimelzon, Fugen Li, Catherine S. Grasso, Mothaffar F. Rimawi, Pavana Anur, Rinath Jeselsohn, Dolores Lopez-Terrada, Dean P. Edwards, Joe W. Gray, Carmine De Angelis, Emporia F Hollingsworth, Resel Pereira, Susan G. Hilsenbeck, Meghana V. Trivedi, Chad J. Creighton, Agostina Nardone, Paul T. Spellman, Nicholas J. Wang, Shixia Huang, Fu, X., Jeselsohn, R., Pereira, R., Hollingsworth, E. F., Creighton, C. J., Li, F., Shea, M., Nardone, A., De Angelis, C., Heiser, L. M., Anur, P., Wang, N., Grasso, C. S., Spellman, P. T., Griffith, O. L., Tsimelzon, A., Gutierrez, C., Huang, S., Edwards, D. P., Trivedi, M. V., Rimawi, M. F., Lopez-Terrada, D., Hilsenbeck, S. G., Gray, J. W., Brown, M., Osborne, C. K., and Schiff, R.
- Subjects
0301 basic medicine ,Hepatocyte Nuclear Factor 3-alpha ,Antineoplastic Agents, Hormonal ,Prognosi ,Estrogen receptor ,Breast Neoplasms ,Biology ,Transcriptome ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,medicine ,Humans ,RNA, Small Interfering ,Gene knockdown ,Multidisciplinary ,Pioneer factor ,Interleukin-8 ,Estrogen Receptor alpha ,medicine.disease ,Prognosis ,Survival Analysis ,Gene Expression Regulation, Neoplastic ,Tamoxifen ,030104 developmental biology ,PNAS Plus ,Drug Resistance, Neoplasm ,Transcriptional reprogramming ,Cancer research ,Female ,Survival Analysi ,FOXA1 ,Estrogen receptor alpha ,Breast Neoplasm ,Endocrine resistance ,Human ,medicine.drug ,Signal Transduction - Abstract
SignificanceOne of the mechanisms of endocrine resistance in estrogen receptor α (ER)-positive (+) breast cancer is the cross-talk between the ER and growth factor receptor pathways leading to altered ER activity and a reprogrammed ER-dependent transcriptome. However, key mediators of this ER-dependent transcriptional reprogramming remain elusive. Here we demonstrate that forkhead box protein A1 (FOXA1) up-regulation via gene amplification or overexpression contributes to endocrine resistance and increased invasiveness phenotypes by altering the ER-dependent transcriptome. We further show that IL-8, one of the top altered FOXA1/ER effectors, plays a key role in mediating these phenotypes and is a potential target to treat ER+/FOXA1-high breast cancer. Our findings provoke a new interplay of FOXA1 in the ER transcriptional program in endocrine-resistant breast cancer.
- Published
- 2016
19. Triple-negative breast cancers: Biomarkers and outcomes
- Author
-
L. Malorni, P. B. Shetty, S. G. Hilsenbeck, M. F. Rimawi, R. M. Elledge, C. De Angelis, C. K. Osborne, DE PLACIDO, SABINO, ARPINO, GRAZIA, Malorni, L., Shetty, P. B., Hilsenbeck, S. G., Rimawi, M. F., Elledge, R. M., De Angelis, C., Osborne, C. K., DE PLACIDO, Sabino, and Arpino, Grazia
- Published
- 2010
20. A web-based data management system for a multicenter international breast cancer oriented blood, tissue, and data biobank.
- Author
-
Margossian, A. L., Saadjian, H., Mira, A., Contreras, A., Rimawi, M. F., Margossian, M. L., Scheurer, M., Osborne, K., and Gutierrez, C.
- Subjects
- *
BIOLOGICAL specimens , *BIOBANKS , *DATA management , *QUALITY assurance , *BREAST cancer , *STATISTICS , *CONFIDENTIAL communications , *INFORMATION retrieval - Abstract
The quality of biospecimens and associated data must be consistent and collected according to standardized methods in order to achieve international harmonization and coordination among biobanking networks. Sharing successful strategies between the Baylor College of Medicine (BCM) Cancer Center and Breast Center Buenos Aires (BCBA) is the driving force for the creation of an Argentinean Breast Cancer oriented Biobank with a shared data management system. Objective: To create a bilingual web-based tool for biospecimen management, inventory, clinical and breast cancer data registration according to international standards of data security quality assurance (ISO 27001). Methods/System Characteristics: The system permits users to enter and retrieve data concerning the collection, storage, quality assurance and distribution of biospecimens, and provides clinical and other patient data for samples while maintaining strict patient confidentiality. It has a multi-tier architecture, developed using Service-Oriented Architecture (SOA) software design principles in .net 4.0, and uses a SQL server database backbone. The system allows multidimensional data analysis trough On Line Analytical Processes (OLAP) and operates with standard web clients, such as Internet Explorer. User Profiles/Access Levels: Clinical staff (e.g., clinical research coordinator, phlebotomist, pathologist, PA, MD) conduct participant and consent registration and preregistration, epidemiological questionnaire data entry, barcode label generation, form printing, and clinical and pathological cancer data entry. * Biospecimen resource staff (e.g., lab technicians) track and store data about biospecimen collection, inventory, tracking and distribution, generate sample barcode labels, and maintain sample allocation and freezer maps for the system. * Scientists have the ability to search for biospecimens and associated data for their own translational research projects. * Breast Cancer Database: Detailed clinical, pathological, treatment and follow-up breast cancer information are captured for each patient, including TNM, biopsy and definitive surgery of primary tumor and events, systemic and radiation treatment, recurrences and metastasis, follow-up. A summary screen provides a snapshot of these data to the viewer. Pathology reports and sample collection forms are attached in each corresponding biopsy or surgery screen for easy viewing and quality assurance. Conclusion: This Biobank Management System was designed by a multidisciplinary team to improve and streamline the workflow for each member of the biobank by: allowing preregistration capabilities before consenting individuals or collecting samples, automatic generation of clinical and sample ID labels, and real-time statistics with certified secure sensible information confidentiality. This system's value is its wide range of uses, from the day-to-day management of a multicenter biobank to the storage of detailed clinical cancerrelated data in a user-friendly and intuitive data entry environment. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
21. Parallel upregulation of Bcl2 and estrogen receptor (ER) expression in HER2+ breast cancer patients treated with neoadjuvant lapatinib.
- Author
-
Giuliano, M., Wang, Y. C., Gutierrez, C., Rimawi, M. F., Chang, J. C., Wang, T., Hilsenbeck, S. G., Trivedi, M. V., Chamness, G. C., Osborne, C. K., and Schiff, R.
- Subjects
- *
HER2 gene , *BREAST cancer , *ESTROGEN receptors , *TUMORS , *LAPATINIB - Abstract
Background: We previously showed in HER2+ models of breast cancer (BC) that potent inhibition of the HER receptor layer can lead to re-expression of estrogen receptor (ER) or activation of the ER pathway. Consequently, the anti-apoptotic ER gene product Bcl2 is upregulated, resulting in enhanced tumor cell survival and treatment resistance. In this study, we investigated whether Bcl2 and ER expression levels are simultaneously increased by neoadjuvant treatment with the dual HER1/2 tyrosine kinase inhibitor lapatinib in HER2+ BC patients. Methods: In a neoadjuvant phase II clinical trial 49 HER2+ BC patients were treated with lapatinib as a single agent for 6 weeks, followed by trastuzumab/docetaxel for 12 weeks before surgery. Tumor specimens were prospectively collected at different time-points during lapatinib treatment (baseline, and weeks 2 and 4). Bcl2, ER, progesterone receptor (PR), total (t) and phosphorylated (p)-HER2, and Ki67 were assessed by immunohistochemistry. Spearman correlation was used to evaluate the association among the biomarkers at baseline, and the correlation of their changes over time. Fisher's Exact test and non-parametric Wilcoxon rank sum test were used respectively to determine if the frequency and the magnitude of Bcl2 expression changes were associated with baseline ER status. Results: 35/49 HER2+ tumor specimens (71%) were available for baseline evaluation of Bcl2 and ER. Of those, 12 (34%) were ER-positive (Allred score 3) and 23 (66%) ER-negative. Baseline Bcl2 expression correlated positively with ER (r = .75; p < .0001) and PR (r = .53; p = .0015), and inversely with t-HER2 (r = -.43; p = .0097). ER baseline expression correlated positively with PR (r = .57; p = .0004), and inversely with t and p-HER2 (r = -.55; p = .0005, and r = -.37; p = .0282, respectively) and Ki67 (r = -.39; p = .0271). Bcl2 changes at week 2 (w2) positively correlated with changes in both ER and PR levels (r = .70; p = .0002 and r = .57; p = .0076, respectively). Additionally, the increase in Bcl2 expression, observed in 9 of the 23 (39%) tumors with tissue available at w2, was significantly more frequent (p = .0147) and of greater magnitude (p = .0001) in ER-pos vs. ER-neg tumors -- 8/9 ER-pos tumors at w2 (including 3 converted from ER-neg by lapatinib) had increased Bcl2, while only 1 of the 14 (7%) ER-neg tumors (at baseline and w2) had increased Bcl2. The expression of ER itself at w2 also increased in 3 out of the 6 (50%) tumors which were originally ER-pos and had tissue available at w2, and in all of them Bcl2 increased in parallel. Of note, the single baseline ER-pos tumor that showed a reduction in ER at w2 had a parallel decrease in Bcl2. Similar observations or trends were found at week 4. Conclusion: Our study suggests that Bcl2 is upregulated as a result of enhanced/restored ER activity upon anti-HER2 therapy with lapatinib in HER2+ tumors. This further supports the use of endocrine along with anti-HER-2 therapy to block this escape pathway which could otherwise cause treatment resistance. In addition, the ER re-expression with lapatinib treatment observed in this study emphasizes the need to re-biopsy HER2+/ER--patients receiving anti-HER2 therapy and to add endocrine therapy if the tumor becomes ER-positive. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. Neratinib and ado-trastuzumab emtansine for pretreated and untreated human epidermal growth factor receptor 2 (HER2)-positive breast cancer brain metastases: Translational Breast Cancer Research Consortium trial 022.
- Author
-
Freedman RA, Heiling HM, Li T, Trapani D, Tayob N, Smith KL, Davis R, Pereslete AM, DeMeo MK, Cotter C, Chen WY, Parsons HA, Santa-Maria CA, Van Poznak C, Moy B, Brufsky AM, Melisko ME, O'Sullivan CC, Ashai N, Rauf Y, Nangia JR, Burns RT, Savoie J, Wolff AC, Winer EP, Rimawi MF, Krop IE, and Lin NU
- Subjects
- Humans, Female, Middle Aged, Adult, Aged, Translational Research, Biomedical, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Ado-Trastuzumab Emtansine administration & dosage, Ado-Trastuzumab Emtansine therapeutic use, Brain Neoplasms secondary, Brain Neoplasms drug therapy, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms genetics, Quinolines administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
Background: Treatment options for human epidermal growth factor receptor 2 (HER2)-positive breast cancer brain metastases (BCBMs) remain limited. We previously reported central nervous system (CNS) activity for neratinib and neratinib-capecitabine. Preclinical data suggest that neratinib may overcome resistance to ado-trastuzumab emtansine (T-DM1) when given in combination. In Translational Breast Cancer Research Consortium (TBCRC) 022's cohort 4, we examined the efficacy of neratinib plus T-DM1 in patients with HER2-positive BCBM., Patients and Methods: In this multicenter, phase II study, patients with measurable HER2-positive BCBM received neratinib 160 mg daily plus T-DM1 3.6 mg/kg intravenously every 21 days in three parallel-enrolling cohorts [cohort 4A-previously untreated BCBM, cohorts 4B and 4C-BCBM progressing after local CNS-directed therapy without (4B) and with (4C) prior exposure to T-DM1]. Cycle 1 diarrheal prophylaxis was required. The primary endpoint was the Response Assessment in Neuro-Oncology-Brain Metastases (RANO-BM) by cohort. The overall survival (OS) and toxicity were also assessed., Results: Between 2018 and 2021, 6, 17, and 21 patients enrolled in cohorts 4A, 4B, and 4C. Enrollment was stopped prematurely for slow accrual. The CNS objective response rate in cohorts 4A, 4B, and 4C was 33.3% [95% confidence interval (CI) 4.3% to 77.7%], 35.3% (95% CI 14.2% to 61.7%), and 28.6% (95% CI 11.3% to 52.2%), respectively; 38.1%-50% experienced stable disease for ≥6 months or response. Diarrhea was the most common grade 3 toxicity (22.7%). The median OS was 30.2 [cohort 4A; 95% CI 21.9-not reached (NR)], 23.3 (cohort 4B; 95% CI 17.6-NR), and 20.9 (cohort 4C; 95% CI 14.9-NR) months., Conclusions: We observed intracranial activity for neratinib plus T-DM1, including those with prior T-DM1 exposure, suggesting synergistic effects with neratinib. Our data provide additional evidence for neratinib-based combinations in patients with HER2-positive BCBM, even those who are heavily pretreated., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
23. Circulating tumor DNA association with residual cancer burden after neoadjuvant chemotherapy in triple-negative breast cancer in TBCRC 030.
- Author
-
Parsons HA, Blewett T, Chu X, Sridhar S, Santos K, Xiong K, Abramson VG, Patel A, Cheng J, Brufsky A, Rhoades J, Force J, Liu R, Traina TA, Carey LA, Rimawi MF, Miller KD, Stearns V, Specht J, Falkson C, Burstein HJ, Wolff AC, Winer EP, Tayob N, Krop IE, Makrigiorgos GM, Golub TR, Mayer EL, and Adalsteinsson VA
- Subjects
- Humans, Female, Neoadjuvant Therapy adverse effects, Neoplasm, Residual genetics, Neoplasm, Residual pathology, Prospective Studies, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local genetics, Circulating Tumor DNA genetics, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Breast Neoplasms etiology
- Abstract
Background: We aimed to examine circulating tumor DNA (ctDNA) and its association with residual cancer burden (RCB) using an ultrasensitive assay in patients with triple-negative breast cancer (TNBC) receiving neoadjuvant chemotherapy., Patients and Methods: We identified responders (RCB 0/1) and matched non-responders (RCB 2/3) from the phase II TBCRC 030 prospective study of neoadjuvant paclitaxel versus cisplatin in TNBC. We collected plasma samples at baseline, 3 weeks and 12 weeks (end of therapy). We created personalized ctDNA assays utilizing MAESTRO mutation enrichment sequencing. We explored associations between ctDNA and RCB status and disease recurrence., Results: Of 139 patients, 68 had complete samples and no additional neoadjuvant chemotherapy. Twenty-two were responders and 19 of those had sufficient tissue for whole-genome sequencing. We identified an additional 19 non-responders for a matched case-control analysis of 38 patients using a MAESTRO ctDNA assay tracking 319-1000 variants (median 1000 variants) to 114 plasma samples from 3 timepoints. Overall, ctDNA positivity was 100% at baseline, 79% at week 3 and 55% at week 12. Median tumor fraction (TFx) was 3.7 × 10
-4 (range 7.9 × 10-7 -4.9 × 10-1 ). TFx decreased 285-fold from baseline to week 3 in responders and 24-fold in non-responders. Week 12 ctDNA clearance correlated with RCB: clearance was observed in 10 of 11 patients with RCB 0, 3 of 8 with RCB 1, 4 of 15 with RCB 2 and 0 of 4 with RCB 3. Among six patients with known recurrence, five had persistent ctDNA at week 12., Conclusions: Neoadjuvant chemotherapy for TNBC reduced ctDNA TFx by 285-fold in responders and 24-fold in non-responders. In 58% (22/38) of patients, ctDNA TFx dropped below the detection level of a commercially available test, emphasizing the need for sensitive tests. Additional studies will determine whether ctDNA-guided approaches can improve outcomes., (Copyright © 2023 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
24. A combinatorial biomarker predicts pathologic complete response to neoadjuvant lapatinib and trastuzumab without chemotherapy in patients with HER2+ breast cancer.
- Author
-
Veeraraghavan J, De Angelis C, Mao R, Wang T, Herrera S, Pavlick AC, Contreras A, Nuciforo P, Mayer IA, Forero A, Nanda R, Goetz MP, Chang JC, Wolff AC, Krop IE, Fuqua SAW, Prat A, Hilsenbeck SG, Weigelt B, Reis-Filho JS, Gutierrez C, Osborne CK, Rimawi MF, and Schiff R
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Class I Phosphatidylinositol 3-Kinases genetics, Class I Phosphatidylinositol 3-Kinases metabolism, Female, Follow-Up Studies, Gene Amplification, Humans, In Situ Hybridization, Fluorescence, Lapatinib administration & dosage, Neoadjuvant Therapy, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Prognosis, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 genetics, Remission Induction, Trastuzumab administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Receptor, ErbB-2 metabolism
- Abstract
Background: HER2-positive (+) breast cancers, defined by HER2 overexpression and/or amplification, are often addicted to HER2 to maintain their malignant phenotype. Yet, some HER2+ tumors do not benefit from anti-HER2 therapy. We hypothesize that HER2 amplification levels and PI3K pathway activation are key determinants of response to HER2-targeted treatments without chemotherapy., Patients and Methods: Baseline HER2+ tumors from patients treated with neoadjuvant lapatinib plus trastuzumab [with endocrine therapy for estrogen receptor (ER)+ tumors] in TBCRC006 (NCT00548184) were evaluated in a central laboratory for HER2 amplification by fluorescence in situ hybridization (FISH) (n = 56). HER2 copy number (CN) and FISH ratios, and PI3K pathway status, defined by PIK3CA mutations or PTEN levels by immunohistochemistry were available for 41 tumors. Results were correlated with pathologic complete response (pCR; no residual invasive tumor in breast)., Results: Thirteen of the 56 patients (23%) achieved pCR. None of the 11 patients with HER2 ratio <4 and/or CN <10 achieved pCR, whereas 13/45 patients (29%) with HER2 ratio ≥4 and/or CN ≥10 attained pCR (P = 0.0513). Of the 18 patients with tumors expressing high PTEN or wild-type (WT) PIK3CA (intact PI3K pathway), 7 (39%) achieved pCR, compared with 1/23 (4%) with PI3K pathway alterations (P = 0.0133). Seven of the 16 patients (44%) with HER2 ratio ≥4 and intact PI3K pathway achieved pCR, whereas only 1/25 (4%) patients not meeting these criteria achieved pCR (P = 0.0031)., Conclusions: Our findings suggest that there is a clinical subtype in breast cancer with high HER2 amplification and intact PI3K pathway that is especially sensitive to HER2-targeted therapies without chemotherapy. A combination of HER2 FISH ratio and PI3K pathway status warrants validation to identify patients who may be treated with HER2-targeted therapy without chemotherapy., (© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.