77 results on '"Rimawi M"'
Search Results
2. Effects of a green tea extract, Polyphenon E, on systemic biomarkers of growth factor signalling in women with hormone receptor‐negative breast cancer
- Author
-
Crew, KD, Ho, KA, Brown, P, Greenlee, H, Bevers, TB, Arun, B, Sneige, N, Hudis, C, McArthur, HL, Chang, J, Rimawi, M, Cornelison, TL, Cardelli, J, Santella, RM, Wang, A, Lippman, SM, and Hershman, DL
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Clinical Trials and Supportive Activities ,Breast Cancer ,Clinical Research ,Prevention ,Nutrition ,Complementary and Integrative Health ,Cancer ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Adult ,Aged ,Biomarkers ,Breast Neoplasms ,Catechin ,Cholesterol ,Female ,Hepatocyte Growth Factor ,Humans ,Intercellular Signaling Peptides and Proteins ,Middle Aged ,Placebos ,Plant Extracts ,Risk Factors ,Signal Transduction ,Tea ,Triglycerides ,Vascular Endothelial Growth Factor A ,breast cancer ,chemoprevention ,tea polyphenols ,Biochemistry and Cell Biology ,Nutrition and Dietetics ,Nutrition & Dietetics ,Clinical sciences ,Nutrition and dietetics - Abstract
BackgroundObservational and experimental data support a potential breast cancer chemopreventive effect of green tea.MethodsWe conducted an ancillary study using archived blood/urine from a phase IB randomised, placebo-controlled dose escalation trial of an oral green tea extract, Polyphenon E (Poly E), in breast cancer patients. Using an adaptive trial design, women with stage I-III breast cancer who completed adjuvant treatment were randomised to Poly E 400 mg (n = 16), 600 mg (n = 11) and 800 mg (n = 3) twice daily or matching placebo (n = 10) for 6 months. Blood and urine collection occurred at baseline, and at 2, 4 and 6 months. Biological endpoints included growth factor [serum hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF)], lipid (serum cholesterol, triglycerides), oxidative damage and inflammatory biomarkers.ResultsFrom July 2007-August 2009, 40 women were enrolled and 34 (26 Poly E, eight placebo) were evaluable for biomarker endpoints. At 2 months, the Poly E group (all dose levels combined) compared to placebo had a significant decrease in mean serum HGF levels (-12.7% versus +6.3%, P = 0.04). This trend persisted at 4 and 6 months but was no longer statistically significant. For the Poly E group, serum VEGF decreased by 11.5% at 2 months (P = 0.02) and 13.9% at 4 months (P = 0.05) but did not differ compared to placebo. At 2 months, there was a trend toward a decrease in serum cholesterol with Poly E (P = 0.08). No significant differences were observed for other biomarkers.ConclusionsOur findings suggest potential mechanistic actions of tea polyphenols in growth factor signalling, angiogenesis and lipid metabolism.
- Published
- 2015
3. 122TiP Phase III study to evaluate the efficacy and safety of GLSI-100 (GP2 + GM-CSF) in breast cancer patients with residual disease or high-risk PCR after both neo-adjuvant and postoperative adjuvant anti-HER2 therapy, Flamingo-01
- Author
-
Patel, S., primary, Thompson, J., additional, Patel, M., additional, Daugherty, F.J., additional, and Rimawi, M., additional
- Published
- 2023
- Full Text
- View/download PDF
4. 43P AKT and estrogen receptor (ER) inhibition potently impairs endocrine resistance (EndoR) in breast cancer (BC)
- Author
-
Nardone, A., primary, Morrison-Thiele, G., additional, Goldstein, A., additional, De Angelis, C., additional, Veeraraghavan, J., additional, Fu, X., additional, Liu, C.C., additional, Wang, T., additional, Shea, M., additional, Cosulich, S., additional, Davies, B., additional, Tsimelzon, A., additional, Huang, S., additional, Chamness, G., additional, Jeselsohn, R., additional, Malorni, L., additional, Rimawi, M., additional, Hilsenbeck, S., additional, Osborne, C.K., additional, and Schiff, R., additional
- Published
- 2023
- Full Text
- View/download PDF
5. 191P Correlation of early change in standardized uptake value (SUV) on positron emission tomography (PET/CT) with recurrence-free survival (RFS) and overall survival (OS) in patients with primary operable HER2-positive breast cancer (TBCRC026)
- Author
-
Hennessy, M.A., primary, Leal, J., additional, Huang, C-Y., additional, Solnes, L., additional, Denbow, R., additional, Abramson, V., additional, Carey, L., additional, Liu, M.C., additional, Rimawi, M., additional, Specht, J., additional, Storniolo, A.M.V., additional, Valero, V., additional, Vaklavas, C., additional, Winer, E., additional, Krop, I., additional, Wolff, A.C., additional, Cimino-Mathews, A., additional, Wahl, R., additional, Stearns, V., additional, and Connolly, R.M., additional
- Published
- 2022
- Full Text
- View/download PDF
6. 328TiP Phase III study of trastuzumab deruxtecan (T-DXd) with or without pertuzumab vs a taxane, trastuzumab and pertuzumab in first-line (1L), human epidermal growth factor receptor 2–positive (HER2+) metastatic breast cancer (mBC): DESTINY-Breast09
- Author
-
Tolaney, S.M., primary, Barroso-Sousa, R., additional, Jiang, Z., additional, Park, Y.H., additional, Rimawi, M., additional, Saura Manich, C., additional, Schneeweiss, A., additional, Toi, M., additional, Yu, T., additional, Shetty, J., additional, Herbolsheimer, P., additional, and Loibl, S., additional
- Published
- 2021
- Full Text
- View/download PDF
7. 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
8. 333P Evaluation of a composite PET/CT and HER2 tissue-based biomarker to predict response to neoadjuvant HER2-directed therapy in early breast cancer (TBCRC026)
- Author
-
Hennessy, M.A., Cimino-Mathews, A., Carter, J., Kachergus, J., Ma, Y., Leal, J., Solnes, L., Abramson, V., Carey, L.A., Rimawi, M., Specht, J., Storniolo, A.M., Vaklavas, C., Wolff, A.C., Wahl, R., Perez, E.A., Huang, C-Y., Stearns, V., Thompson, A., and Connolly, R.M.
- Published
- 2023
- Full Text
- View/download PDF
9. 353TiP Phase III study to evaluate the efficacy and safety of GLSI-100 (GP2 + GM-CSF) in breast cancer patients with residual disease or high-risk PCR after both neo-adjuvant and postoperative adjuvant anti-HER2 therapy, Flamingo-01
- Author
-
Patel, S., Thompson, J., Patel, M., Daugherty, F.J., and Rimawi, M.
- Published
- 2023
- Full Text
- View/download PDF
10. ERBB2 mRNA as a predictor in HER2-positive (HER2+)/hormone receptor-positive (HR+) metastatic breast cancer (BC) treated with HER2 blockade in combination with endocrine therapy (ET): A retrospective analysis of the ALTERNATIVE and SOLTI-PAMELA trials
- Author
-
Chic, N., primary, Pascual, T., additional, Brasó-Maristany, F., additional, Villagrasa Gonzalez, P., additional, Pare Brunet, L., additional, Schettini, F., additional, Conte, B., additional, Adamo, B., additional, Vidal, M., additional, Muñoz, M., additional, Martínez, O., additional, Gonzalez-Farre, B., additional, Cortés, J., additional, Llombart-Cussac, A., additional, Rodrik-Outmezguine, V., additional, Izquierdo Delso, M.A., additional, Schiff, R., additional, Osborne, C.K., additional, Rimawi, M., additional, and Prat, A., additional
- Published
- 2019
- Full Text
- View/download PDF
11. Abstract ES1-3: Advanced HER2-positive breast cancer: Overcoming treatment resistance
- Author
-
Rimawi, M, primary
- Published
- 2019
- Full Text
- View/download PDF
12. Abstract GS3-02: PALLET: A neoadjuvant study to compare the clinical and antiproliferative effects of letrozole with and without palbociclib
- Author
-
Dowsett, M, primary, Jacobs, S, additional, Johnston, S, additional, Bliss, J, additional, Wheatley, D, additional, Holcombe, C, additional, Stein, R, additional, McIntosh, S, additional, Barry, P, additional, Dolling, D, additional, Snowdon, C, additional, Perry, S, additional, Batten, L, additional, Dodson, A, additional, Martins, V, additional, Modi, A, additional, Cornman, C, additional, Puhalla, S, additional, Wolmark, N, additional, Julian, T, additional, Pogue-Geile, K, additional, Robidoux, A, additional, Provencher, L, additional, Boileau, JF, additional, Shalaby, I, additional, Thirlwell, M, additional, Fisher, K, additional, Huang Bartlett, C, additional, Koehler, M, additional, Osborne, K, additional, and Rimawi, M, additional
- Published
- 2019
- Full Text
- View/download PDF
13. Abstract P3-11-01: Treatment patterns for young women with HR+/HER2- metastatic breast cancer in the United States in the era of CDK 4/6 inhibitors
- Author
-
Burstein, HJ, primary, Mayer, EL, additional, DeMichele, A, additional, Harnett, J, additional, Mardekian, J, additional, McRoy, L, additional, Huang Bartlett, C, additional, Koehler, M, additional, and Fahed Rimawi, M, additional
- Published
- 2018
- Full Text
- View/download PDF
14. 119P - ERBB2 mRNA as a predictor in HER2-positive (HER2+)/hormone receptor-positive (HR+) metastatic breast cancer (BC) treated with HER2 blockade in combination with endocrine therapy (ET): A retrospective analysis of the ALTERNATIVE and SOLTI-PAMELA trials
- Author
-
Chic, N., Pascual, T., Brasó-Maristany, F., Villagrasa Gonzalez, P., Pare Brunet, L., Schettini, F., Conte, B., Adamo, B., Vidal, M., Muñoz, M., Martínez, O., Gonzalez-Farre, B., Cortés, J., Llombart-Cussac, A., Rodrik-Outmezguine, V., Izquierdo Delso, M.A., Schiff, R., Osborne, C.K., Rimawi, M., and Prat, A.
- Published
- 2019
- Full Text
- View/download PDF
15. Abstract OT3-02-08: Scalp cooling alopecia prevention trial (SCALP) for patients with early stage breast cancer
- Author
-
Nangia, JR, primary, Wang, T, additional, Rude, M, additional, Osborne, C, additional, Papish, S, additional, Abraham, J, additional, Holmes, F, additional, Savin, M, additional, Paxman, R, additional, Hilsenbeck, SG, additional, Osborne, CK, additional, and Rimawi, M, additional
- Published
- 2016
- Full Text
- View/download PDF
16. Abstract P4-01-11: Limits of [18F]-FLT PET as a clinical biomarker of proliferation in breast cancer
- Author
-
Schmidt, EV, primary, Blackman, S, additional, Iannone, R, additional, Senderak, ET, additional, Railkar, RA, additional, Evelhoch, JL, additional, Mozley, PD, additional, Perez, EA, additional, McDonough, M, additional, Rimawi, M, additional, Tolaney, S, additional, Kim, S-B, additional, and Chung, H-C, additional
- Published
- 2013
- Full Text
- View/download PDF
17. Abstract P5-18-24: Population pharmacokinetics of trastuzumab emtansine, a HER2-targeted antibody-drug conjugate, in patients with HER2-positive metastatic breast cancer: clinical implications of the effect of various covariates
- Author
-
Lu, D, primary, Girish, S, additional, Gao, Y, additional, Wang, B, additional, Yi, J-H, additional, Guardino, E, additional, Samant, M, additional, Cobleigh, M, additional, Rimawi, M, additional, Conte, P, additional, and Jin, J, additional
- Published
- 2012
- Full Text
- View/download PDF
18. S07 Overcoming resistance to endocrine therapies: Multiple interventions to reach the goal
- Author
-
Schiff, R., primary, Osborne, K., additional, Rimawi, M., additional, and Malorni, L., additional
- Published
- 2011
- Full Text
- View/download PDF
19. Targeting Intrinsically-Resistant Breast Cancer Stem Cells with Gamma-Secretase Inhibitors.
- Author
-
Chang, J., primary, Landis, M., additional, Schott, A., additional, Pavlick, A., additional, Dobrolecki, L., additional, Korkaya, H., additional, Zhang, X., additional, Froehlich, A., additional, Rodriguez, A., additional, Rimawi, M., additional, Wicha, M., additional, Lewis, M., additional, and Hayes, D., additional
- Published
- 2009
- Full Text
- View/download PDF
20. A BRCA1-Like, 25-Gene Assay Predicts for Anthracycline-Chemosensitivity in Sporadic Triple-Negative Breast Cancer.
- Author
-
Rodriguez, A., primary, Rimawi, M., additional, Wu, M., additional, Dave, B., additional, Wong, H., additional, Landis, M., additional, Cairo, M., additional, Pavlick, A., additional, Froehlich, A., additional, Chamness, G., additional, Hilsenbeck, S., additional, Lewis, M., additional, Osborne, C., additional, and Chang, J., additional
- Published
- 2009
- Full Text
- View/download PDF
21. PI3 kinase activation and response to trastuzumab or lapatinib in HER-2 overexpressing locally advanced breast cancer (LABC).
- Author
-
Migliaccio, I, primary, Gutierrez, M, additional, Wu, M, additional, Wong, H, additional, Pavlick, A, additional, Hilsenbeck, SG, additional, Horlings, HM, additional, Rimawi, M, additional, Berns, K, additional, Bernards, R, additional, Osborne, C, additional, Arteaga, CL, additional, and Chang, JC, additional
- Published
- 2009
- Full Text
- View/download PDF
22. BRCA1 gene expression signature predicts for anthracycline-chemosensitivity in triple-negative breast cancer.
- Author
-
Rodriguez, AA, primary, Makris, A, additional, Harrison, MK, additional, Ostler, PJ, additional, Froehlich, A, additional, Pavlick, A, additional, Wong, H, additional, Tsimelzon, A, additional, Sexton, K, additional, Hilsenbeck, SG, additional, Lewis, MT, additional, Rimawi, M, additional, Osborne, CK, additional, and Chang, JC, additional
- Published
- 2009
- Full Text
- View/download PDF
23. Combination antiHER therapeutics — efficacy of alternative dosing regimens in ER+/HER2+ breast cancer xenografts.
- Author
-
Wiechmann, LS, primary, Soliz, R, additional, Hilsenbeck, S, additional, Rimawi, M, additional, Osborne, CK, additional, and Schiff, R, additional
- Published
- 2009
- Full Text
- View/download PDF
24. Treatment of Human Epidermal Growth Factor Receptor 2-Overexpressing Breast Cancer Xenografts With Multiagent HER-Targeted Therapy
- Author
-
Arpino, G., primary, Gutierrez, C., additional, Weiss, H., additional, Rimawi, M., additional, Massarweh, S., additional, Bharwani, L., additional, De Placido, S., additional, Osborne, C. K., additional, and Schiff, R., additional
- Published
- 2007
- Full Text
- View/download PDF
25. 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
26. 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
27. 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
28. 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
29. 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
30. 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
31. 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
32. 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
33. 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
34. Circulating tumor cell investigation in breast cancer patient-derived xenograft models by automated immunofluorescence staining, image acquisition, and single cell retrieval and analysis
- Author
-
Chandandeep Nagi, Hariprasad Thangavel, Mario Giuliano, Lacey E. Dobrolecki, Agostina Nardone, Carmine De Angelis, Meghana V. Trivedi, Michael T. Lewis, Rachel Schiff, Jackie L. Stilwell, C. Kent Osborne, Debashish Sahay, Sina Hedayatpour, Raksha Bhat, Eric P. Kaldjian, Mothaffar F. Rimawi, Arturo Ramirez, Ramirez, A. B., Bhat, R., Sahay, D., De Angelis, C., Thangavel, H., Hedayatpour, S., Dobrolecki, L. E., Nardone, A., Giuliano, M., KHALIL RODRIGUEZ, Nagi, Rimawi, M., Osborne, C. K., Lewis, M. T., Stilwell, J. L., Kaldjian, E. P., Schiff, R., and Trivedi, M. V.
- Subjects
0301 basic medicine ,Cancer Research ,Cell Separation ,Mice, SCID ,Metastasis ,Antineoplastic Agent ,Mice ,0302 clinical medicine ,Circulating tumor cell ,Breast cancer ,Single-cell analysis ,Class I Phosphatidylinositol 3-Kinase ,medicine.diagnostic_test ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Neoplastic Cells, Circulating ,3. Good health ,Patient-derived xenografts ,Oncology ,030220 oncology & carcinogenesis ,Single-cell analysi ,Immunohistochemistry ,Keratins ,Female ,Single-Cell Analysis ,Breast Neoplasm ,Research Article ,Human ,Class I Phosphatidylinositol 3-Kinases ,Antineoplastic Agents ,Breast Neoplasms ,Immunofluorescence ,lcsh:RC254-282 ,03 medical and health sciences ,Patient-derived xenograft ,Cell Line, Tumor ,Genetics ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Chemotherapy ,Cluster of differentiation ,business.industry ,Animal ,Circulating tumor cells ,Leukocyte Common Antigen ,Sequence Analysis, DNA ,medicine.disease ,030104 developmental biology ,Keratin ,Cancer cell ,Mutation ,Cancer research ,Leukocyte Common Antigens ,business ,Neoplasm Transplantation - Abstract
Background Breast cancer patient-derived xenograft (BC-PDX) models represent a continuous and reproducible source of circulating tumor cells (CTCs) for studying their role in tumor biology and metastasis. We have previously shown the utility of BC-PDX models in the study of CTCs by immunohistochemistry (IHC) on serial paraffin sections and manual microscopic identification of cytokeratin-positive cells, a method that is both low-throughput and labor-intensive. We therefore aimed to identify and characterize CTCs from small volume mouse blood samples and examined its practical workflow in a study of BC-PDX mice treated with chemotherapy using an automated imaging platform, the AccuCyte®–CyteFinder® system. Methods CTC analysis was conducted using blood from non-tumor bearing SCID/Beige mice spiked with human breast cancer cells, BC-PDX-bearing mice, and BC-PDX mice treated with vehicle or chemotherapeutic agent(s). After red blood cell lysis, nucleated cells were mixed with transfer solution, processed onto microscope slides, and stained by immunofluorescence. The CyteFinder automated scanning microscope was used to identify CTCs, defined as nucleated cells that were human cytokeratin-positive, and mouse CD45-negative. Disaggregated primary BC-PDX tumors and lung metastatic nodules were processed using the same immunostaining protocol. Collective expression of breast cancer cell surface markers (EpCAM, EGFR, and HER2) using a cocktail of target-specific antibodies was assessed. CTCs and disaggregated tumor cells were individually retrieved from slides using the CytePicker® module for sequence analysis of a BC-PDX tumor-specific PIK3CA mutation. Results The recovery rate of human cancer cells spiked into murine blood was 83 ± 12%. CTC detection was not significantly different from the IHC method. One-third of CTCs did not stain positive for cell surface markers. A PIK3CA T1035A mutation present in a BC-PDX tumor was confirmed in isolated single CTCs and cells from dissociated metastatic nodules after whole genome amplification and sequencing. CTC evaluation could be simply implemented into a preclinical PDX therapeutic study setting with substantial improvements in workflow over the IHC method. Conclusions Analysis of small volume blood samples from BC-PDX-bearing mice using the AccuCyte–CyteFinder system allows investigation of the role of CTCs in tumor biology and metastasis independent of surface marker expression. Electronic supplementary material The online version of this article (10.1186/s12885-019-5382-1) contains supplementary material, which is available to authorized users.
- Published
- 2019
35. 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
36. 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
37. 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
38. Population pharmacokinetics of trastuzumab emtansine, a HER2-targeted antibody-drug conjugate, in patients with HER2-positive metastatic breast cancer: clinical implications of the effect of various covariates.
- Author
-
Lu, D., Girish, S., Gao, Y., Wang, B., Yi, J.-H., Guardino, E., Samant, M., Cobleigh, M., Rimawi, M., Conte, P., and Jin, J.
- Subjects
- *
TRASTUZUMAB , *PHARMACOKINETICS , *PHARMACOLOGY , *SERUM , *SULFIDES , *DRUGS - Abstract
Background: Trastuzumab emtansine (T-DM1) is a HER2-targeted antibody-drug conjugate composed of the humanized monoclonal antibody trastuzumab, the potent cytotoxic agent DM1 (a microtubule inhibitor), and a stable thioether linker. To estimate typical pharmacokinetic (PK) parameter values and interpatient variability, a population PK model for T-DM1 was previously developed from 1 phase 1 (0.3 to 4.8 mg/kg in qw or q3w regimens) and 2 phase 2 (3.6 mg/kg q3w) trials (Gupta, J Clin Pharmacol2012). The model reported here has been updated with additional data from 2 randomized trials (phase 2 TDM4450g and phase 3 EMILIA, 3.6 mg/kg q3w). Another phase 2 trial (TDM4688g) was used for external validation of the model. The effect of demographic and pathophysiological covariates on the PK of T-DM1 was explored to better understand the clinical factors that might affect exposure and clinical outcome for individual patients. Methods: For the current analysis, 9934 T-DM1 serum concentration-time data points from 671 patients were simultaneously fitted using NONMEM® software. T-DM1 concentration-time data to date are best described using a 2-compartment linear model. All relevant and plausible covariates likely to have an effect on T-DM1 systemic exposure, or likely to have clinical relevance, were explored for possible correlation with the key T-DM1 PK parameters of clearance (CL) and central volume of distribution (Vc). These covariates include those related to demographics, renal and hepatic function, disease status, and treatment history. Results: The estimated CL for T-DM1 is 0.68 L/day, Vc is 3.13 L, and the terminal half-life is 3.94 days. Interindividual variability (IIV) of the base model is 25.6% and 17.5% for CL and Vc, respectively. Patients with greater body weight, sum of longest dimension of target lesions, serum concentration of shed HER2 extracellular domain, and aspartate aminotransferase concentrations, as well as patients with lower serum albumin and baseline trastuzumab concentrations, have statistically faster CL. Patients with greater body weight also have statistically larger Vc. Incorporation of these covariates (P<0.001 by likelihood ratio test) decreased IIV of CL and Vc to 19.1 % and 11.7%, respectively. All covariates together explain 44.4% and 55.8% of IIV in CL and Vc, respectively. The model sensitivity analysis suggests that a patient with a statistically significant PK covariate value at the 5th or 95th percentile of the population will have a <20% difference in cumulative exposure (as represented by area under the T-DM1 concentration-time curve) compared with a typical patient with a median covariate value. Conclusions: A relatively small IIV for the estimated T-DM1 PK parameters of CL and Vc was observed. None of the evaluated covariates had a clinically meaningful magnitude of effect on T-DM1 exposure (<20% difference for patients with 5th and 95th percentiles vs patients with median value of covariates) that would justify a further dose adjustment. The body weight-based dose of 3.6 mg/kg q3w without further correction for other factors is considered appropriate in ongoing clinical trials. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
39. 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
40. 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
41. Calibrating Observational Health Record Data Against a Randomized Trial.
- Author
-
Merola D, Campbell U, Lenis D, Schneeweiss S, Wang S, Madsen A, Carrigan G, Chia V, Ovbiosa OE, Pinheiro S, Pace N, Bruno A, Stewart M, Khosla S, Zhang Y, Rimawi M, Hendricks-Sturrup R, Huang J, Taylor A, Jiao X, Becnel L, McRoy L, Eckert J, Rodriguez C, Lunacsek O, Harvey R, Greshock J, Sarsour K, Belli A, Wang CK, Fernandes L, Chen J, San Francisco B, Sangli C, Natanzon Y, Chan KA, Dhopeshwarkar N, Shapiro M, Wasserman A, Quinn J, Rees M, Robinson T, Taylor B, and Rider JR
- Subjects
- Humans, Male, Female, Middle Aged, Retrospective Studies, Aged, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung mortality, Antibodies, Monoclonal, Humanized therapeutic use, Calibration, Pilot Projects, Electronic Health Records statistics & numerical data, Randomized Controlled Trials as Topic, Lung Neoplasms drug therapy, Lung Neoplasms mortality
- Abstract
Importance: The conditions required for health record data sources to accurately assess treatment effectiveness remain unclear. Emulation of randomized clinical trials (RCTs) with health record data and subsequent calibration of the results can help elucidate this., Objective: To pilot an emulation of the KEYNOTE-189 RCT using a commercially available electronic health record (EHR) data source., Design, Setting, and Participants: This retrospective cohort study used an EHR database spanning from April 2007 to February 2023. Follow-up began on treatment initiation and proceeded until an outcome event, loss to follow-up, end of data, or end of study period (640 days). The population-based cohort was ascertained from EHRs provided by 52 health systems across the US. Eligibility criteria were defined as closely as possible to the benchmark RCT. Patients with non-small cell lung cancer initiating first-line treatment for metastatic disease were included. Patients with evidence of squamous non-small cell lung cancer, primary nonlung malignant neoplasms, or identified EGFR/ALK variations were excluded. Data were analyzed from June to October 2023., Exposures: Initiation of first-line pembrolizumab and chemotherapy and chemotherapy alone. Chemotherapy in both groups was defined as a combination of pemetrexed and platinum-based (carboplatin or cisplatin) therapy., Main Outcomes and Measures: Outcomes of interest were 12-month survival probability and mortality hazard ratio (HR)., Results: A total of 1854 patients (mean [SD] age, 63.7 [9.6] years; 971 [52.4%] men) were eligible, including 589 patients who initiated pembrolizumab and chemotherapy and 1265 patients who initiated chemotherapy only. The cohort included 364 Black patients (19.6%) and 1445 White patients (77.9%). The 12-month survival probabilities were 0.60 (95% CI, 0.54-0.65) in the pembrolizumab group and 0.58 (95% CI, 0.55-0.62) in the chemotherapy-only group, compared with 0.69 (95% CI, 0.64-0.74) in the KEYNOTE-189 pembrolizumab group and 0.49 (95% CI, 0.42-0.56) in the KEYNOTE-189 chemotherapy-only group. The mortality HR was 0.95 (95% CI, 0.78-1.16), compared with 0.49 (95% CI, 0.38-0.64) in the KEYNOTE-189 RCT., Conclusions and Relevance: In this cohort study piloting an RCT emulation, results were incongruous with the benchmark trial. Differences in patient treatment and data capture between the RCT and EHR populations, confounding by indication, treatment crossover, and accuracy of captured diagnoses may explain these findings. Future feasibility assessments will require data sources to have important oncology-specific measures curated.
- Published
- 2024
- Full Text
- View/download PDF
42. Effect of cross-platform gene-expression, computational methods on breast cancer subtyping in PALOMA-2 and PALLET studies.
- Author
-
Cheang MCU, Rimawi M, Johnston S, Jacobs SA, Bliss J, Pogue-Geile K, Kilburn L, Zhu Z, Schuster EF, Xiao H, Swaim L, Deng S, Lu DR, Gauthier E, Tursi J, Slamon DJ, Rugo HS, Finn RS, and Liu Y
- Abstract
Intrinsic breast cancer molecular subtyping (IBCMS) provides significant prognostic information for patients with breast cancer and helps determine treatment. This study compared IBCMS methods on various gene-expression platforms in PALOMA-2 and PALLET trials. PALOMA-2 tumor samples were profiled using EdgeSeq and nanostring and subtyped with AIMS, PAM50, and research-use-only (ruo)Prosigna. PALLET tumor biopsies were profiled using mRNA sequencing and subtyped with AIMS and PAM50. In PALOMA-2 (n = 222), a 54% agreement was observed between results from AIMS and gold-standard ruoProsigna, with AIMS assigning 67% basal-like to HER2-enriched. In PALLET (n = 224), a 69% agreement was observed between results from PAM50 and AIMS. Different IBCMS methods may lead to different results and could misguide treatment selection; hence, a standardized clinical PAM50 assay and computational approach should be used.Trial number: NCT01740427., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
43. Metformin: A Dual-Role Player in Cancer Treatment and Prevention.
- Author
-
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, and Aljada A
- Subjects
- Humans, Cell Proliferation, Chemotherapy, Adjuvant, Hyperplasia, Metformin pharmacology, Metformin therapeutic use, Diabetes Mellitus, Type 2 drug therapy, Neoplasms drug therapy
- Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
44. Novel TLR7 hemizygous variant in post-COVID-19 neurological deterioration: a case report with literature review.
- Author
-
Noor Eddin A, Al-Rimawi M, Peer-Zada F, Hundallah K, and Alhashem A
- Abstract
The neurological complications of coronavirus disease 2019 (COVID-19) can range from simple tremors and dystonia to features of encephalopathy. Toll-like receptor 7 (TLR7) belongs to a family of innate immune receptors responsible for viral RNA detection (such as SARS-CoV-2) and immune response initiation. TLR7 loss of function variants have been previously reported as genetic risk factors for severe COVID-19 infection in young patients with no comorbidities. In this case, we report a pediatric patient who developed severe long-term neurological deterioration following his COVID-19 infection. Presenting first to the clinic with episodic dystonia and finger spasticity, the patient's condition rapidly deteriorated with a significant drop in the Glasgow Coma Scale (GCS). Despite improvement following initial treatment with rituximab and intravenous immunoglobulin, the patient's symptoms relapsed, and GCS further dropped to 3/15. Serial brain magnetic resonance imaging scans revealed diffuse parenchymal atrophy, ventricular enlargement, and spinal cord thickening. Autoimmune investigations were negative but clinical whole genome sequencing prioritized four gene variants, the most significant of which was a novel frameshift null variant of the X chromosomal TLR7 gene (c.1386_1389dup, p.[His464Ilefs*7]). This case illustrates a role for TLR7 in long-term COVID-19 complications and highlights that TLR7 deficiency in the future may be addressed as a therapeutic measure., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Noor Eddin, Al-Rimawi, Peer-Zada, Hundallah and Alhashem.)
- Published
- 2023
- Full Text
- View/download PDF
45. Correlation of SUV on Early Interim PET with Recurrence-Free Survival and Overall Survival in Primary Operable HER2-Positive Breast Cancer (the TBCRC026 Trial).
- Author
-
Hennessy MA, Leal JP, Huang CY, Solnes LB, Denbow R, Abramson VG, Carey LA, Liu MC, Rimawi M, Specht J, Storniolo AM, Valero V, Vaklavas C, Winer EP, Krop IE, Wolff AC, Cimino-Mathews A, Wahl RL, Stearns V, and Connolly RM
- Subjects
- Humans, Female, Fluorodeoxyglucose F18, Positron Emission Tomography Computed Tomography, Treatment Outcome, Receptor, ErbB-2 metabolism, Trastuzumab, Positron-Emission Tomography, Neoadjuvant Therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Breast Neoplasms metabolism
- Abstract
Predictive biomarkers of response to human epidermal growth factor receptor 2 (HER2)-directed therapy are essential to inform treatment decisions. The TBCRC026 trial reported that early declines in tumor SUVs corrected for lean body mass (SUL
max ) on18 F-FDG PET/CT predicted a pathologic complete response (pCR) to HER2 therapy with neoadjuvant trastuzumab and pertuzumab (HP) without chemotherapy in estrogen receptor (ER)-negative, HER2-positive breast cancer. We hypothesized that18 F-FDG PET/CT SULmax parameters would predict recurrence-free survival (RFS) and overall survival (OS). Methods: Patients with stage II/III ER-negative, HER2-positive breast cancer received neoadjuvant HP ( n = 88). pCR after HP alone was 22% (18/83), additional nonstudy neoadjuvant therapy was administered in 28% (25/88), and the majority received adjuvant therapy per physician discretion.18 F-FDG PET/CT was performed at baseline and at cycle 1, day 15 (C1D15). RFS and OS were summarized using the Kaplan-Meier method and compared between subgroups using logrank tests. Associations between18 F-FDG PET/CT (≥40% decline in SULmax between baseline and C1D15, or C1D15 SULmax ≤ 3) and pCR were evaluated using Cox regressions, where likelihood ratio CIs were reported because of the small numbers of events. Results: Median follow-up was 53.7 mo (83/88 evaluable), with 6 deaths and 14 RFS events. Estimated RFS and OS at 3 y was 84% (95% CI, 76%-92%) and 92% (95% CI, 87%-98%), respectively. A C1D15 SULmax of 3 or less was associated with improved RFS (hazard ratio [HR], 0.36; 95% CI, 0.11-1.05; P = 0.06) and OS (HR, 0.14; 95% CI, 0.01-0.85; P = 0.03), the latter statistically significant. The association of an SULmax decline of at least 40% (achieved in 59%) with RFS and OS did not reach statistical significance. pCR was associated with improved RFS (HR, 0.25; 95% CI, 0.01-1.24; P = 0.10) but did not reach statistical significance. Conclusion: For the first time, we report a potential association between a C1D15 SULmax of 3 or less on18 F-FDG PET/CT and RFS and OS outcomes in patients with ER-negative, HER2-positive breast cancer receiving neoadjuvant HP alone. If confirmed in future studies, this imaging-based biomarker may facilitate early individualization of therapy., (© 2023 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2023
- Full Text
- View/download PDF
46. 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
47. Cerebrospinal fluid microRNAs as potential biomarkers in Alzheimer's disease.
- Author
-
Noor Eddin A, Hamsho K, Adi G, Al-Rimawi M, Alfuwais M, Abdul Rab S, Alkattan K, and Yaqinuddin A
- Abstract
Alzheimer's disease (AD) is the leading form of dementia worldwide, but its early detection and diagnosis remain a challenge. MicroRNAs (miRNAs) are a group of small endogenous RNA molecules that regulate mRNA expression. Recent evidence suggests miRNAs play an important role in the five major hallmarks of AD pathophysiology: amyloidogenesis, tauopathy, neuroinflammation, synaptic dysfunction, and neuronal death. Compared to traditional biomarkers of AD, miRNAs display a greater degree of stability in cerebrospinal fluid. Moreover, aberrant changes in miRNA expression can be measured over time to monitor and guide patient treatment. Specific miRNA profiles and combinations may also be used to distinguish AD subjects from normal controls and other causes of dementia. Because of these properties, miRNAs are now being considered as promising and potential biomarkers of AD. This review comprehensively summarizes the diagnostic potential and regulatory roles miRNAs play in AD., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Noor Eddin, Hamsho, Adi, Al-Rimawi, Alfuwais, Abdul Rab, Alkattan and Yaqinuddin.)
- Published
- 2023
- Full Text
- View/download PDF
48. Toward Practical Integration of Omic and Imaging Data in Co-Clinical Trials.
- Author
-
Alkim E, Dowst H, DiCarlo J, Dobrolecki LE, Hernández-Herrera A, Hormuth DA 2nd, Liao Y, McOwiti A, Pautler R, Rimawi M, Roark A, Srinivasan RR, Virostko J, Zhang B, Zheng F, Rubin DL, Yankeelov TE, and Lewis MT
- Subjects
- Humans, Magnetic Resonance Imaging, Image Processing, Computer-Assisted, Triple Negative Breast Neoplasms pathology
- Abstract
Co-clinical trials are the concurrent or sequential evaluation of therapeutics in both patients clinically and patient-derived xenografts (PDX) pre-clinically, in a manner designed to match the pharmacokinetics and pharmacodynamics of the agent(s) used. The primary goal is to determine the degree to which PDX cohort responses recapitulate patient cohort responses at the phenotypic and molecular levels, such that pre-clinical and clinical trials can inform one another. A major issue is how to manage, integrate, and analyze the abundance of data generated across both spatial and temporal scales, as well as across species. To address this issue, we are developing MIRACCL (molecular and imaging response analysis of co-clinical trials), a web-based analytical tool. For prototyping, we simulated data for a co-clinical trial in "triple-negative" breast cancer (TNBC) by pairing pre- (T0) and on-treatment (T1) magnetic resonance imaging (MRI) from the I-SPY2 trial, as well as PDX-based T0 and T1 MRI. Baseline (T0) and on-treatment (T1) RNA expression data were also simulated for TNBC and PDX. Image features derived from both datasets were cross-referenced to omic data to evaluate MIRACCL functionality for correlating and displaying MRI-based changes in tumor size, vascularity, and cellularity with changes in mRNA expression as a function of treatment.
- Published
- 2023
- Full Text
- View/download PDF
49. Neutrophil extracellular traps in central nervous system pathologies: A mini review.
- Author
-
Shafqat A, Noor Eddin A, Adi G, Al-Rimawi M, Abdul Rab S, Abu-Shaar M, Adi K, Alkattan K, and Yaqinuddin A
- Abstract
Neutrophils are the first cells to be recruited to sites of acute inflammation and contribute to host defense through phagocytosis, degranulation and neutrophil extracellular traps (NETs). Neutrophils are rarely found in the brain because of the highly selective blood-brain barrier (BBB). However, several diseases disrupt the BBB and cause neuroinflammation. In this regard, neutrophils and NETs have been visualized in the brain after various insults, including traumatic (traumatic brain injury and spinal cord injury), infectious (bacterial meningitis), vascular (ischemic stroke), autoimmune (systemic lupus erythematosus), neurodegenerative (multiple sclerosis and Alzheimer's disease), and neoplastic (glioma) causes. Significantly, preventing neutrophil trafficking into the central nervous system or NET production in these diseases alleviates brain pathology and improves neurocognitive outcomes. This review summarizes the major studies on the contribution of NETs to central nervous system (CNS) disorders., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Shafqat, Noor Eddin, Adi, Al-Rimawi, Abdul Rab, Abu-Shaar, Adi, Alkattan and Yaqinuddin.)
- Published
- 2023
- Full Text
- View/download PDF
50. Multi-antigen-targeted T-cell therapy to treat patients with relapsed/refractory breast cancer.
- Author
-
Hoyos V, Vasileiou S, Kuvalekar M, Watanabe A, Tzannou I, Velazquez Y, French-Kim M, Leung W, Lulla S, Robertson C, Foreman C, Wang T, Bulsara S, Lapteva N, Grilley B, Ellis M, Osborne CK, Coscio A, Nangia J, Heslop HE, Rooney CM, Vera JF, Lulla P, Rimawi M, and Leen AM
- Abstract
Purpose: Adoptively transferred, ex vivo expanded multi-antigen-targeted T cells (multiTAA-T) represent a new, potentially effective, and nontoxic therapeutic approach for patients with breast cancer (BC). In this first-in-human trial, we investigated the safety and clinical effects of administering multiTAA T cells targeting the tumor-expressed antigens, Survivin, NY-ESO-1, MAGE-A4, SSX2, and PRAME, to patients with relapsed/refractory/metastatic BC., Materials and Methods: MultiTAA T-cell products were generated from the peripheral blood of heavily pre-treated patients with metastatic or locally recurrent unresectable BC of all subtypes and infused at a fixed dose level of 2 × 10
7 /m2 . Patients received two infusions of cells 4 weeks apart and safety and clinical activity were determined. Cells were administered in an outpatient setting and without prior lymphodepleting chemotherapy., Results: All patients had estrogen receptor/progesterone receptor positive BC, with one patient also having human epidermal growth factor receptor 2-positive. There were no treatment-related toxicities and the infusions were well tolerated. Of the 10 heavily pre-treated patients enrolled and infused with multiTAA T cells, nine had disease progression while one patient with 10 lines of prior therapies experienced prolonged (5 months) disease stabilization that was associated with the in vivo expansion and persistence of T cells directed against the targeted antigens. Furthermore, antigen spreading and the endogenous activation of T cells directed against a spectrum of non-targeted tumor antigens were observed in 7/10 patients post-multiTAA infusion., Conclusion: MultiTAA T cells were well tolerated and induced disease stabilization in a patient with refractory BC. This was associated with in vivo T-cell expansion, persistence, and antigen spreading. Future directions of this approach may include additional strategies to enhance the therapeutic benefit of multiTAA T cells in patients with BC., Competing Interests: Competing Interests: S.V., M.K., and Y.V. are consultants to AlloVir. V.H. holds Marker Therapeutics and AlloVir stock. N.L. is a consultant to Tessa Therapeutics. J.F.V. is a cofounder and equity holder in AlloVir and Marker Therapeutics and an employee of Marker Therapeutics, which aspires to commercialize the described approach. B.J.G. owns QBRegulatory Consulting which has consulting agreements with Tessa Therapeutics, Marker Therapeutics, LOKON, and AlloVir. H.E.H. is a co-founder with equity in Allovir and Marker Therapeutics, has served on advisory boards for Tessa Therapeutics, Kiadis, Novartis, Gilead Biosciences, Fresh Wind Biotechnologies and GSK, and received research support from Kuur Therapeutics and Tessa Therapeutics. C.M.R. has Stock and Other Ownership Interests with Coya, Bluebird Bio, Tessa Therapeutics, Marker Therapeutics, AlloVir, Walking Fish, Allogene Therapeutics, Memgen, Kuur Therapeutics, Bellicum Pharmaceuticals, TScan Therapeutics, Abintus Bio; Consulting or Advisory Role with Abintus Bio, Adaptimmune, Brooklyn Immunotherapeutic, Onk Therapeutics, Tessa Therapeutics, Memgen, Torque, Walking Fish Therapeutics, TScan Therapeutics, Marker Therapeutics, Turnstone Bio; and receives research funding from Kuur Therapeutics. A.M.L is a co-founder and equity holder for AlloVir and Marker Therapeutics and a consultant to AlloVir. P.L. is a member of the advisory board for Karyopharm. J.N. receives research support from Paxman Coolers Ltd. M.R. is a consultant to AstraZeneca, Macrogenics, Seagen and Novartis and receives research support from Pfizer. The remaining authors have no competing financial interests to disclose., (© The Author(s), 2022.)- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.