Phase I Study and Cell-Free DNA Analysis of T-DM1 and Metronomic Temozolomide for Secondary Prevention of HER2-Positive Breast Cancer Brain Metastases

Sarah Jenkins, Wei Zhang, Seth M Steinberg, Darryl Nousome, Nicole Houston, Xiaolin Wu, Terri S Armstrong, Eric Burton, Dee Dee Smart, Ritu Shah, Cody J Peer, Brett Mozarsky, Oluwatobi Arisa, William D Figg, Tito R Mendoza, Elizabeth Vera, Priscilla Brastianos, Scott Carter, Mark R Gilbert, Carey K Anders, Roisín M Connolly, Carol Tweed, Karen L Smith, Imran Khan, Stanley Lipkowitz, Patricia S Steeg, Alexandra S Zimmer, Sarah Jenkins, Wei Zhang, Seth M Steinberg, Darryl Nousome, Nicole Houston, Xiaolin Wu, Terri S Armstrong, Eric Burton, Dee Dee Smart, Ritu Shah, Cody J Peer, Brett Mozarsky, Oluwatobi Arisa, William D Figg, Tito R Mendoza, Elizabeth Vera, Priscilla Brastianos, Scott Carter, Mark R Gilbert, Carey K Anders, Roisín M Connolly, Carol Tweed, Karen L Smith, Imran Khan, Stanley Lipkowitz, Patricia S Steeg, Alexandra S Zimmer

Abstract

Purpose: Preclinical data showed that prophylactic, low-dose temozolomide (TMZ) significantly prevented breast cancer brain metastasis. We present results of a phase I trial combining T-DM1 with TMZ for the prevention of additional brain metastases after previous occurrence and local treatment in patients with HER2+ breast cancer.

Patients and methods: Eligible patients had HER2+ breast cancer with brain metastases and were within 12 weeks of whole brain radiation therapy (WBRT), stereotactic radiosurgery, and/or surgery. Standard doses of T-DM1 were administered intravenously every 21 days (3.6 mg/kg) and TMZ was given orally daily in a 3+3 phase I dose escalation design at 30, 40, or 50 mg/m2, continuously. DLT period was one 21-day cycle. Primary endpoint was safety and recommended phase II dose. Symptom questionnaires, brain MRI, and systemic CT scans were performed every 6 weeks. Cell-free DNA sequencing was performed on patients' plasma and CSF.

Results: Twelve women enrolled, nine (75%) with prior SRS therapy and three (25%) with prior WBRT. Grade 3 or 4 AEs included thrombocytopenia (1/12), neutropenia (1/12), lymphopenia (6/12), and decreased CD4 (6/12), requiring pentamidine for Pneumocystis jirovecii pneumonia prophylaxis. No DLT was observed. Four patients on the highest TMZ dose underwent dose reductions. At trial entry, 6 of 12 patients had tumor mutations in CSF, indicating ongoing metastatic colonization despite a clear MRI. Median follow-up on study was 9.6 m (2.8-33.9); only 2 patients developed new parenchymal brain metastases. Tumor mutations varied with patient outcome.

Conclusions: Metronomic TMZ in combination with standard dose T-DM1 shows low-grade toxicity and potential activity in secondary prevention of HER2+ brain metastases.

Conflict of interest statement

SJ, WZ, SMS, NH, TSA, EB, DS, RS, CJP, BM; OA; WDF; TRM; EV; MRG; IK, SL, DN, XW declare no potential conflicts of interest.

PSS receives research funding from Daichi-Sanyo/AstraZeneca.

PKB has consulted for Tesaro, Angiochem, Genentech-Roche, ElevateBio, Eli Lilly, SK Life Sciences, Advise Connect Inspire (ACI), Pfizer, Voyager Therapeutics, Sintetica, and Dantari, received institutional research funding (to MGH) from Merck, Mirati, Eli Lilly, BMS, and Pfizer, and has received honoraria from Merck, Pfizer, and Genentech-Roche

CKA has received research funding PUMA, Lilly, Merck, Seattle Genetics, Nektar, Tesaro, G1-Therapeutics, ZION, Novartis, Pfizer; compensated consultant role for Genentech (1/2019-), Eisai (1/2019-), IPSEN (2/2019 - ), Seattle Genetics (11/15/2019 - 11/15/2020); Astra Zeneca (3/2020 -), Novartis (5/2020 - 5/2022), Immunomedics (10/1/2020 - 9/22/2021), Elucida (9/2020), Athenex (2/2021 - 2/2023); and royalties from UpToDate, Jones and Bartlet.

RC has received an unrestricted educational grant from Pfizer, and research funding to institution for clinical trials from MSD Ireland, Pfizer, Daichii Sankyo, and Astra Zeneca.

CT have received consultant and speaker fees from Seagen.

KLS has received research funding (to institution) from Pfizer and her husband has stock in Abbott Labs and Abbvie.

ASZ has received speaker fees from Medscape.

©2023 American Association for Cancer Research.

Figures

Figure 1.. Time on Study and Outcomes.
Figure 1.. Time on Study and Outcomes.
Patients received a standard regimen on 3.6 mg/m2 of T-DM1 iv. q21days and one of three indicated doses of oral TMZ (30, 40 or 50 mg/m2, dose levels 1-3, respectively), daily for each 21-day cycle. Patient number is indicated to the left. Initial local treatment and status as of 10/21 data cutoff is shown. Only two patients developed new parenchymal brain metastases over a median of 9.6 months. SRS, stereotactic radiation surgery prior to entering the study; WBRT, whole brain radition therapy prior to entering the study; PD, progressive systemic disease, LMD, leptomeningeal disease. # >20 lesions at baseline. * Radionecrosis confirmed after patient off study for suspected PD that was not confirmed.
Figure 2.. Whole-exome sequencing of plasma and…
Figure 2.. Whole-exome sequencing of plasma and CSF cfDNA samples from HER2+ breast cancer patients.
(A) Schematic of phase I study with regard to samples collected. The sample collection time points of four plasma samples and two CSF samples are labelled as “+”. (B-D) Identification of genomic alternations in the plasma and CSF cfDNA samples by Patient Number (Fig. 1). For each sample genomic alterations were compared to known variants for cancer-related genes in the MSK-IMPACT panel (B), and main pathogenic/likely pathogenic for all diseases in the ClinVar database (C). The potential driver mutations were identified using the OncodriveCLUST method (D). P, plasma; CSF, cerebrospinal fluid. The bar graph above each panel totals the number of mutations detected in each cfDNA sample. On the left, the percentage of cfDNA reads with mutations in the genes is listed. On the right, the distribution of mutation type per gene is indicated. On the bottom of each panel, blue lines identify two patients who developed new parenchymal brain metastases during this trial, while red lines identify four long-term survivors (>12 months without a new parenchymal brain metastasis).
Figure 3.. Mutation patterns for patients with…
Figure 3.. Mutation patterns for patients with new brain metastases versus long-term responders.
Potential relationship between mutations in plasma and CSF cfDNA throughout the study, and the patients’ clinical outcome. Two patients who developed new brain metastases during this trial are listed as “New Brain Metastasis”, while four patients who remained free of a new parenchymal brain metastasis for at least 12 months were identified as “Long-term survivors” (see Figure 1). The type of mutations is indicated by a colored circle.

References

    1. Barnholtz-Sloan JS, Sloan AE, Davis FG, Vigneau FD, Lai P, Sawaya RE. Incidence proportions of brain metastases in patients diagnosed (1973 to 2001) in the metropolitan Detroit cancer surveillance system. Journal of Clinical Oncology 2004;22:2865–72.
    1. Bendell J, Domchek S, Burstein H, Harris L, Younger J, Kuter I, et al. Central Nervous System Metastases in Women who Receive Trastuzumab-Based Therapy for Metastatic Breast Carcinoma. Cancer 2003;97:2972–7.
    1. Clayton A, Danson S, Jolly S, Ryder W, Burt P, Stewart A, et al. Incidence of cerebral metastases in patients treated with trastuzumab for metastatic breast cancer. British Journal of Cancer 2004;91:639–43.
    1. Olson EM, Abdel-Rasoul M, Maly J, Wu CS, Lin NU, Shapiro CL. Incidence and risk of central nervous system metastases as site of first recurrence in patients with HER2-positive breast cancer treated with adjuvant trastuzumab. Annals of Oncology 2013;24:1526–33.
    1. Kased N, Binder DK, McDermott MW, Nakamura JL, Huang K, Berger MS, et al. Gamma Knife Radiosurgery For Brain Metastases From Primary Breast Cancer. International Journal of Radiation Oncology Biology Physics 2009;75:1132–40.
    1. Bachelot T, Romieu G, Campone M, Dieras V, Cropet C, Dalenc F, et al. Lapatinib plus capecitabine in patients with previously untreated brain metastases from HER2-positive metastatic breast cancer (LANDSCAPE): a single-group phase 2 study. Lancet Oncol 2013;14:64–71.
    1. Freedman RA, Gelman RS, Anders CK, Melisko ME, Parsons HA, Cropp AM, et al. TBCRC 022: A Phase II Trial of Neratinib and Capecitabine for Patients With Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer and Brain Metastases. Journal of Clinical Oncology 2019;37:1081.
    1. Achrol AS, Rennert RC, Anders C, Soffietti R, Ahluwalia MS, Nayak L, et al. Brain metastases. Nature Reviews Disease Primers 2019;5:5.
    1. Auperin A, Arriagada R, Pignon JP, Le Pechoux C, Gregor A, Stephens RJ, et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. N Engl J Med 1999;341:476–84.
    1. van Grinsven EE, Nagtegaal SHJ, Verhoeff JJC, van Zandvoort MJE. The Impact of Stereotactic or Whole Brain Radiotherapy on Neurocognitive Functioning in Adult Patients with Brain Metastases: A Systematic Review and Meta-Analysis. Oncology Research and Treatment 2021;44:622–36.
    1. Brown PD, Asher AL, Ballman KV, Farace E, Cerhan JH, Anderson SK, et al. NCCTG N0574 (Alliance): A phase III randomized trial of whole brain radiation therapy (WBRT) in addition to radiosurgery (SRS) in patients with 1 to 3 brain metastases. Journal of Clinical Oncology 2015;33.
    1. Steeg PS. PERSPECTIVE The right trials. Nature 2012;485:S58–S9.
    1. Trudeau ME, Crump M, Charpentier D, Yelle L, Bordeleau L, Matthews S, et al. Temozolomide in metastatic breast cancer (MBC): A phase II trial of the National Cancer Institute of Canada-Clinical Trials Group (NCIC-CTG). Annals of Oncology 2006;17:952–6.
    1. Addeo R, Caraglia M, Faiola V, Capasso E, Vincenzi B, Montella L, et al. Concomitant treatment of brain metastasis with Whole Brain Radiotherapy WBRT and Temozolomide TMZ is active and improves Quality of Life. Bmc Cancer 2007;7.
    1. Addeo R, De Rosa C, Faiola V, Leo L, Cennamo G, Montella L, et al. Phase 2 Trial of Temozolomide Using Protracted Low-dose and Whole-brain Radiotherapy for Nonsmall Cell Lung Cancer and Breast Cancer Patients With Brain Metastases. Cancer 2008;113:2524–31.
    1. Addeo R, Sperlongano P, Montella L, Vincenzi B, Carraturo M, Iodice P, et al. Protracted low dose of oral vinorelbine and temozolomide with whole-brain radiotherapy in the treatment for breast cancer patients with brain metastases. Cancer Chemotherapy and Pharmacology 2012;70:603–9.
    1. de Azambuja E, Zardavas D, Lemort M, Rossari J, Moulin C, Buttice A, et al. Phase I trial combining temozolomide plus lapatinib for the treatment of brain metastases in patients with HER2-positive metastatic breast cancer: the LAPTEM trial. Annals of Oncology 2013;24:2985–9.
    1. Iwamoto FM, Omuro AM, Raizer JJ, Nolan CP, Hormigo A, Lassman AB, et al. A phase II trial of vinorelbine and intensive temozolomide for patients with recurrent or progressive brain metastases. Journal of Neuro-Oncology 2008;87:85–90.
    1. Palmieri D, Duchnowska R, Woditschka S, Hua E, Qian YZ, Biernat W, et al. Profound Prevention of Experimental Brain Metastases of Breast Cancer by Temozolomide in an MGMT-Dependent Manner. Clinical Cancer Research 2014;20:2727–39.
    1. Omuro A, Chan TA, Abrey LE, Khasraw M, Reiner AS, Kaley TJ, et al. Phase II trial of continuous low-dose temozolomide for patients with recurrent malignant glioma. Neuro-Oncology 2013;15:242–50.
    1. Kong DS, Lee JI, Kim JH, Kim ST, Kim WS, Suh YL, et al. Phase II trial of low-dose continuous (metronomic) treatment of temozolomide for recurrent glioblastoma. Neuro-Oncology 2010;12:289–96.
    1. Zimmer AS, Steinberg SM, Smart DD, Gilbert MR, Armstrong TS, Burton E, et al. Temozolomide in secondary prevention of HER2-positive breast cancer brain metastases. Future Oncology 2020;16.
    1. El Mubarak MA, Stylos EK, Chatziathanasiadou MV, Danika C, Alexiou GA, Tsekeris P, et al. Development and validation of simple step protein precipitation UHPLC-MS/MS methods for quantitation of temozolomide in cancer patient plasma samples. Journal of Pharmaceutical and Biomedical Analysis 2019;162:164–70.
    1. A Goyal HK, K Lee, R garg, S Yun et al. . Ultra-Fast Next Generation Human Genome Sequencing Data Processing Using DRAGENTM Bio-IT Processor for Precision Medicine. Open Journal of Genetics 2017;7:9–19.
    1. Brock CS, Newlands ES, Wedge SR, Bower M, Evans H, Colquhoun L, et al. Phase I trial of temozolomide using an extended continuous oral schedule. Cancer Research 1998;58:4363–7.
    1. Dhodapkar M, Rubin J, Reid JM, Burch PA, Pitot HC, Buckner JC, et al. Phase I trial of temozolomide (NSC 362856) in patients with advanced cancer. Clinical Cancer Research 1997;3:1093–100.
    1. Lu D, Girish S, Gao YY, Wang B, Yi JH, Guardino E, et al. Population pharmacokinetics of trastuzumab emtansine (T-DM1), a HER2-targeted antibody-drug conjugate, in patients with HER2-positive metastatic breast cancer: clinical implications of the effect of covariates. Cancer Chemotherapy and Pharmacology 2014;74:399–410.
    1. Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): A Hybridization Capture-Based Next-Generation Sequencing Clinical Assay for Solid Tumor Molecular Oncology. J Mol Diagn 2015;17:251–64.
    1. Landrum MJ, Lee JM, Benson M, Brown GR, Chao C, Chitipiralla S, et al. ClinVar: improving access to variant interpretations and supporting evidence. Nucleic Acids Res 2018;46:D1062–d7.
    1. Mularoni L, Sabarinathan R, Deu-Pons J, Gonzalez-Perez A, López-Bigas N. OncodriveFML: a general framework to identify coding and non-coding regions with cancer driver mutations. Genome Biology 2016;17:128.
    1. Brastianos PK, Carter SL, Santagata S, Cahill DP, Taylor-Weiner A, Jones RT, et al. Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets. Cancer Discovery 2015;5:1164–77.
    1. Montemurro F, Delaloge S, Barrios CH, Wuerstlein R, Anton A, Brain E, et al. Trastuzumab emtansine (T-DM1) in patients with HER2-positive metastatic breast cancer and brain metastases: exploratory final analysis of cohort 1 from KAMILLA, a single-arm phase IIIb clinical trial. Annals of Oncology 2020;31:1350–8.
    1. Baker SD, Wirth M, Statkevich P, Reidenberg P, Alton K, Sartorius SE, et al. Absorption, metabolism, and excretion of C-14-temozolomide following oral administration to patients with advanced cancer. Clinical Cancer Research 1999;5:309–17.
    1. Ostermann S, Csajka C, Buclin T, Leyvraz S, Lejeune F, Decosterd LA, et al. Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients. Clinical Cancer Research 2004;10:3728–36.
    1. Brown PD, Gondi V, Pugh S, Tome WA, Wefel JS, Armstrong TS, et al. Hippocampal Avoidance During Whole-Brain Radiotherapy Plus Memantine for Patients With Brain Metastases: Phase III Trial NRG Oncology CC001. Journal of Clinical Oncology 2020;38:1019–29.
    1. Cheok SK, Narayan A, Arnal-Estape A, Gettinger S, Goldberg SB, Kluger HM, et al. Tumor DNR Mutations From Intraparenchymal Brain Metastases Are Detectable in CSF. Jco Precision Oncology 2021;5:163–72.
    1. Li YM, Pan WY, Connolly ID, Reddy S, Nagpal S, Quake S, et al. Tumor DNA in cerebral spinal fluid reflects clinical course in a patient with melanoma leptomeningeal brain metastases. Journal of Neuro-Oncology 2016;128:93–100.
    1. Cheng X, Zhao JX, Dong F, Cao XC. ARID1A Mutation in Metastatic Breast Cancer: A Potential Therapeutic Target. Frontiers in Oncology 2021;11.
    1. Niedermaier B, Sak A, Zernickel E, Xu S, Groneberg M, Stuschke M. Targeting ARID1A-mutant colorectal cancer: depletion of ARID1B increases radiosensitivity and modulates DNA damage response. Scientific Reports 2019;9.
    1. Helming KC, Wang XF, Wilson BG, Vazquez F, Haswell JR, Manchester HE, et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Nature Medicine 2014;20:251–4.
    1. Helming K, Wang XF, Wilson B, Vazquez F, Haswell J, Manchester H, et al. ARID1B is a specific vulnerability in ARID1A-mutant cancers. Cancer Research 2014;74.
    1. Chen BF, Chan WY. The de novo DNA methyltransferase DNMT3A in development and cancer. Epigenetics 2014;9:669–77.
    1. Karlow JA, Devarakonda S, Xing XY, Jang HS, Govindan R, Watson M, et al. Developmental Pathways Are Epigenetically Reprogrammed during Lung Cancer Brain Metastasis. Cancer Research 2022;82:2692–703.
    1. Marzese DM, Witz IP, Kelly DF, Hoon DSB. Epigenomic landscape of melanoma progression to brain metastasis: unexplored therapeutic alternatives. Epigenomics 2015;7:1303–11.
    1. Khier S, Lohan L. Kinetics of circulating cell-free DNA for biomedical applications: critical appraisal of the literature. Future Science Oa 2018;4.

Source: PubMed

3
Abonnere