Trastuzumab Emtansine (T-DM1)

Total Page:16

File Type:pdf, Size:1020Kb

Trastuzumab Emtansine (T-DM1) Published OnlineFirst September 11, 2018; DOI: 10.1158/1078-0432.CCR-18-1590 Research Article Clinical Cancer Research Trastuzumab Emtansine (T-DM1) in Patients with Previously Treated HER2-Overexpressing Metastatic Non–Small Cell Lung Cancer: Efficacy, Safety, and Biomarkers Solange Peters1, Rolf Stahel2, Lukas Bubendorf3, Philip Bonomi4, Augusto Villegas5, Dariusz M. Kowalski6, Christina S. Baik7, Dolores Isla8, Javier De Castro Carpeno9, Pilar Garrido10, Achim Rittmeyer11, Marcello Tiseo12, Christoph Meyenberg13, Sanne de Haas14, Lisa H. Lam15, Michael W. Lu15, and Thomas E. Stinchcombe16 Abstract Purpose: HER2-targeted therapy is not standard of care Results: Forty-nine patients received T-DM1 (29 IHC 2þ, for HER2-positive non–small cell lung cancer (NSCLC). This 20 IHC 3þ). No treatment responses were observed in the phase II study investigated efficacy and safety of the HER2- IHC 2þ cohort. Four partial responses were observed in the targeted antibody–drug conjugate trastuzumab emtansine IHC 3þ cohort (ORR, 20%; 95% confidence interval, 5.7%– (T-DM1) in patients with previously treated advanced 43.7%). Clinical benefit rates were 7% and 30% in the IHC HER2-overexpressing NSCLC. 2þ and 3þ cohorts, respectively. Response duration for the Patients and Methods: Eligible patients had HER2-over- responders was 2.9, 7.3, 8.3, and 10.8 months. Median expressing NSCLC (centrally tested IHC) and received progression-free survival and overall survival were similar previous platinum-based chemotherapy and targeted between cohorts. Three of 4 responders had HER2 gene therapy in the case of EGFR mutation or ALK gene amplification. No new safety signals were observed. rearrangement. Patients were divided into cohorts based Conclusions: T-DM1 showed a signal of activity in patients on HER2 IHC (2þ,3þ). All patients received T-DM1 3.6 with HER2-overexpressing (IHC 3þ) advanced NSCLC. Addi- mg/kg intravenously every 3 weeks until disease progres- tional investigation into HER2 pathway alterations is needed sion or unacceptable toxicity. The primary endpoint was to refine the target population for T-DM1 in NSCLC; however, investigator-determined overall response rate (ORR) HER2 IHC as a single parameter was an insufficient predictive using RECIST v1.1. biomarker. Clin Cancer Res; 1–9. Ó2018 AACR. 1Oncology Department, Lausanne University Hospital, Lausanne, Introduction Switzerland. 2Cancer Center Zurich,€ University Hospital of Zurich,€ Zurich,€ – 3 The development of targeted therapy for non small cell lung Switzerland. Institute of Pathology, University Hospital Basel, Basel, fi Switzerland. 4Section of Medical Oncology, Rush University Medical Center, cancer (NSCLC) with speci c molecular alterations such as anaplastic lymphoma kinase ALK EGFR Chicago, Illinois. 5Florida Cancer Specialists and Research Institute, Fleming ( ) rearrangements or muta- Island, Florida. 6Center of Oncology, Maria Skłodowska Curie Memorial tions represents a tremendous advance (1, 2). Current research Cancer Centre, Warsaw, Poland. 7SeattleCancerCenterAlliance,University is focused on the identification and development of targeted 8 of Washington, Seattle, Washington. Medical Oncology Section, Hospital therapy for additional molecular subtypes. HER2 is overexpress- Clinico Universitario Lozano Blesa, Zaragoza, Spain. 9Medical Oncology 10 ed on the surface of multiple tumor cell types, including NSCLC Section, Hospital Universitario La Paz, Madrid, Spain. Medical Oncology – Department, Hospital Universitario Ramon y Cajal, Madrid, Spain. (3 5). Survival data meta-analyses show HER2 overexpression is 11Department of Thoracic Oncology, Fachklinik fur€ Lungenerkrankungen, associated with poor prognosis in lung cancer (3, 6). There are Immenhausen, Germany. 12Medical Oncology Unit, Azienda Ospedaliero- currently no standard therapies targeting the HER2 pathway in Universitaria di Parma, Parma, Italy. 13Biostatistics, F. Hoffmann-La Roche, NSCLC, whereas HER2-targeted therapies are standard for breast 14 Basel, Switzerland. Oncology Biomarker Development, F. Hoffmann-La and gastric cancer. In breast and gastric cancer, HER2 overexpres- Roche, Basel, Switzerland. 15Product Development Oncology, Genentech, fi 16 sion generally occurs in the context of gene ampli cation, whereas Inc., South San Francisco, California. Duke Cancer Institute, Duke University fi School of Medicine, Durham, North Carolina. discordance between IHC overexpression and gene ampli cation is observed in NSCLC (7À10). Instead, increased HER2 expres- Note: Supplementary data for this article are available at Clinical Cancer sion in NSCLC may result from upregulated transcriptional/ Research Online (http://clincancerres.aacrjournals.org/). posttranscriptional mechanisms (7, 9). Corresponding Author: Solange Peters, Lausanne University Hospital, Lausanne The reported prevalence of "HER2-positive" NSCLC varies 1011, Switzerland. Phone: 41795560192; E-mail: [email protected] because previous studies have used different definitions and doi: 10.1158/1078-0432.CCR-18-1590 testing methods (ISH and IHC), with HER2 positivity defined Ó2018 American Association for Cancer Research. by IHC 2þ/3þ staining reported in 13% to 20%, IHC 3þ staining www.aacrjournals.org OF1 Downloaded from clincancerres.aacrjournals.org on September 23, 2021. © 2018 American Association for Cancer Research. Published OnlineFirst September 11, 2018; DOI: 10.1158/1078-0432.CCR-18-1590 Peters et al. to increased HER2 transcription, independent of HER2 gene Translational Relevance amplification or mutation (7À10). To date, there are no clinical There are no standard therapies targeting HER2 in non– data suggesting T-DM1, in contrast to trastuzumab or a HER2 small cell lung cancer (NSCLC); however, HER2-targeted tyrosine kinase inhibitor (TKI), would have a stronger effect in therapies are standard in breast and gastric cancer. Results HER2-mutated or HER2-ampli fied NSCLC. Although amplifi- from this phase II study indicate a signal of activity of the cation and mutations are considered potential driver onco- HER2-targeted antibody–drug conjugate trastuzumab genes, we hypothesize that targeting HER2 overexpression will emtansine (T-DM1) in patients with IHC 3þ HER2-positive address a potentially larger patient population that may be NSCLC. T-DM1 was tolerable in NSCLC, and safety was more relevant taking into account the mechanism of action of similar to findings from prior trials. An exploratory bio- T-DM1, targeting the HER2 extracellular component. marker analysis showed that, of the 4 responding patients, 3 had HER2-amplified tumors and 2 had HER2 mutations. Additional investigation into HER2 oncogenic modifica- Materials and Methods tions, including HER2 overexpression, amplification, or Study design and patients mutation may help refine a patient population likely to In this multicenter, single-arm, clinical trial (trial registration benefit from treatment with T-DM1. Of importance, HER2 NCT02289833), eligible patients were aged 18 years with IHC as a single parameter was an insufficient predictive HER2-positive (IHC 2þ or 3þ) locally advanced or metastatic biomarker to select patients with most benefitfromT-DM1. NSCLC previously treated with 1 prior platinum-based chemo- Further trials should refine the target population for HER2- therapeutic regimen. Patients with EGFR-mutated or ALK gene– targeted therapies in NSCLC. rearranged NSCLC were eligible if they had also experienced disease progression following treatment with prior targeted ther- apy or if they were intolerant to such treatment. Archived tumor specimens from previously collected tissue were centrally and prospectively tested for HER2 status (Ventana in 2% to 6%, and HER2 gene amplification by ISH in 2% to 4% Pathway HER2 (4B5) IHC assay; Ventana Medical Systems, Inc.). (8). HER2 gene mutation, another HER2 alteration in NSCLC, has HER2 overexpression was evaluated by IHC, the gold standard a frequency of 1% to 4% (8). HER2 gene mutations and ampli- for HER2 assessment in breast and gastric cancer. If archival tissue fications show limited co-occurrence in NSCLC (8, 10). The was unavailable for HER2 testing, patients could have a newly absence of correlation between HER2 overexpression, amplifica- collected biopsy specimen tested. Based on results from central tion, or mutation suggests 3 biologically distinct NSCLC subtypes, testing, patients with a HER2 status of IHC 2þ (defined as weak- leaving the question of which subtypes will be most effectively to-moderate complete, basolateral, or lateral membranous reac- treated with HER2-targeted therapy. tivity in 10% of tumor cells) or IHC 3þ (defined as strong Despite observation of these 3 alterations in NSCLC, the role of complete, basolateral, or lateral membranous reactivity in 10% these abnormalities as therapeutic biomarkers remains unde- of tumor cells) were eligible. fined. Few data sets report on the use of HER2-targeted therapies A retrospective exploratory biomarker analysis was conducted in patients with HER2-positive (11, 12) or -mutated (13–16) if sufficient tissue was available following IHC testing. In this NSCLC. These studies were small, and some included patients analysis, HER2 gene amplification (HER2/CEP17 gene ratio 2) treated concurrently with chemotherapy and a HER2-targeting was also assessed by ISH (similar to breast and gastric cancers). agent. Of note, most trials do not assess presence of all 3 types of HER2 mRNA expression levels were measured by quantitative HER2 alterations, and tend to use significantly divergent
Recommended publications
  • (AZD6244) in an in Vivo Model of Childhood Astrocytoma
    Author Manuscript Published OnlineFirst on October 16, 2013; DOI: 10.1158/1078-0432.CCR-13-0842 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Development, Characterization, and Reversal of Acquired Resistance to the MEK1 Inhibitor Selumetinib (AZD6244) in an In Vivo Model of Childhood Astrocytoma Hemant K. Bid1, Aaron Kibler1, Doris A. Phelps1, Sagymbek Manap1, Linlin Xiao1, Jiayuh Lin1, David Capper2, Duane Oswald1, Brian Geier1, Mariko DeWire1,5, Paul D. Smith3, Raushan T. Kurmasheva1, Xiaokui Mo4, Soledad Fernandez4, and Peter J. Houghton1*. 1Center for Childhood Cancer & Blood Diseases, Nationwide Children’s Hospital, Columbus, OH 43205 2Institut of Pathology, Department Neuropathology, Ruprecht-Karls University and Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany 3Astrazeneca Ltd., Oncology iMed, Macclesfield, U.K. 4Center for Biostatistics, The Ohio State University, Columbus, OH 43221 5 Present address: Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 Correspondence to Peter J. Houghton, Ph.D. Center for Childhood Cancer & Blood Diseases Nationwide Children’s Hospital 700 Children’s Drive Columbus, OH 43205 Ph: 614-355-2633 Fx: 614-355-2792 [email protected] Running head: Acquired resistance to MEK Inhibition in astrocytoma models. Conflict of Interest Statement: The authors consider that there is no actual or perceived conflict of interest. Dr. Paul D. Smith is an employee of Astrazeneca. 1 Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2013 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 16, 2013; DOI: 10.1158/1078-0432.CCR-13-0842 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]
  • Could Hbx Protein Expression Affect Signal Pathway Inhibition by Gefitinib Or Selumetinib, a MEK Inhibitor, in Hepatocellular Carcinoma Cell Lines?
    ORIGINAL ARTICLE Oncology & Hematology DOI: 10.3346/jkms.2011.26.2.214 • J Korean Med Sci 2011; 26: 214-221 Could HBx Protein Expression Affect Signal Pathway Inhibition by Gefitinib or Selumetinib, a MEK Inhibitor, in Hepatocellular Carcinoma Cell Lines? Yoon Kyung Park1, Kang Mo Kim1, Hepatitis B virus X (HBx) protein has been known to play an important role in development Young-Joo Lee2, Ki-Hun Kim2, of hepatocellular carcinoma (HCC). The aim of this study is to find out whether HBx Sung-Gyu Lee2, Danbi Lee1, protein expression affects antiproliferative effect of an epidermal growth factor receptor- Ju Hyun Shim1, Young-Suk Lim1, tyrosine kinase (EGFR-TK) inhibitor and a MEK inhibitor in HepG2 and Huh-7 cell lines. We 1 1 Han Chu Lee , Young-Hwa Chung , established HepG2 and Huh-7 cells transfected stably with HBx gene. HBx protein 1 1 Yung Sang Lee , and Dong Jin Suh expression increased pERK and pAkt expression as well as β-catenin activity in both cells. Departments of 1Internal Medicine and 2Surgery, Gefitinib (EGFR-TK inhibitor) inhibited pERK and pAkt expression andβ -catenin activity in Asan Medical Center, University of Ulsan College of both cells. Selumetinib (MEK inhibitor) reduced pERK level and β-catenin activity but pAkt Medicine, Seoul, Korea expression was rather elevated by selumetinib in these cells. Reduction of pERK levels was much stronger with selumetinib than gefitinib in both cells. The antiproliferative efficacy Received: 19 July 2010 Accepted: 2 November 2010 of selumetinib was more potent than that of gefitinib. However, the antiproliferative effect of gefitinib, as well as selumetinib, was not different between cell lines with or Address for Correspondence: without HBx expression.
    [Show full text]
  • Press Release
    Press Release Daiichi Sankyo and AstraZeneca Announce Global Development and Commercialization Collaboration for Daiichi Sankyo’s HER2 Targeting Antibody Drug Conjugate [Fam-] Trastuzumab Deruxtecan (DS-8201) Collaboration combines Daiichi Sankyo’s scientific and technological excellence with AstraZeneca’s global experience and resources in oncology to accelerate and expand the potential of [fam-] trastuzumab deruxtecan as monotherapy and combination therapy across a spectrum of HER2 expressing cancers AstraZeneca to pay Daiichi Sankyo up to $6.90 billion in total consideration, including $1.35 billion upfront payment and up to an additional $5.55 billion contingent upon achievement of future regulatory and sales milestones as well as other contingencies Companies to share equally development and commercialization costs as well as profits worldwide from [fam-] trastuzumab deruxtecan with Daiichi Sankyo maintaining exclusive rights in Japan Daiichi Sankyo is expected to book sales in U.S., certain countries in Europe, and certain other markets where Daiichi Sankyo has affiliates; AstraZeneca is expected to book sales in all other markets worldwide, including China, Australia, Canada and Russia Tokyo, Munich and Basking Ridge, NJ – (March 28, 2019) – Daiichi Sankyo Company, Limited (hereafter, Daiichi Sankyo) announced today that it has entered into a global development and commercialization agreement with AstraZeneca for Daiichi Sankyo’s lead antibody drug conjugate (ADC), [fam-] trastuzumab deruxtecan (DS-8201), currently in pivotal development for multiple HER2 expressing cancers including breast and gastric cancer, and additional development in non-small cell lung and colorectal cancer. Daiichi Sankyo and AstraZeneca will jointly develop and commercialize [fam-] trastuzumab deruxtecan as a monotherapy or a combination therapy worldwide, except in Japan where Daiichi Sankyo will maintain exclusive rights.
    [Show full text]
  • Phase I/II Study Evaluating the Safety and Clinical Efficacy of Temsirolimus and Bevacizumab in Patients with Chemotherapy Refra
    Investigational New Drugs (2019) 37:331–337 https://doi.org/10.1007/s10637-018-0687-5 PHASE II STUDIES Phase I/II study evaluating the safety and clinical efficacy of temsirolimus and bevacizumab in patients with chemotherapy refractory metastatic castration-resistant prostate cancer Pedro C. Barata1 & Matthew Cooney2 & Prateek Mendiratta 2 & Ruby Gupta3 & Robert Dreicer4 & Jorge A. Garcia3 Received: 25 September 2018 /Accepted: 16 October 2018 /Published online: 7 November 2018 # Springer Science+Business Media, LLC, part of Springer Nature 2018 Summary Background Mammalian target of rapamycin (mTOR) pathway and angiogenesis through vascular endothelial growth factor (VEGF) have been shown to play important roles in prostate cancer progression. Preclinical data in prostate cancer has suggested the potential additive effect dual inhibition of VEGF and mTOR pathways. In this phase I/II trial we assessed the safety and efficacy of bevacizumab in combination with temsirolimus for the treatment of men with metastatic castration-resistant prostate cancer (mCRPC). Methods In the phase I portion, eligible patients received temsirolimus (20 mg or 25 mg IV weekly) in combination with a fixed dose of IV bevacizumab (10 mg/kg every 2 weeks). The primary endpoint for the phase II portion was objective response measured by either PSA or RECIST criteria. Exploratory endpoints included changes in circulating tumor cells (CTC) and their correlation with PSA response to treatment. Results Twenty-one patients, median age 64 (53–82), with pre- treatment PSA of 205.3 (11.1–1801.0), previously treated with a median of 2 (0–5) lines of therapy for mCRPC received the combination of temsirolimus weekly at 20 mg (n =4)or25mg(n = 17) with bevacizumab 10 mg/kg every 2 weeks (n =21).
    [Show full text]
  • Pediatric Patients with Refractory Central Nervous System Tumors: Experiences of a Clinical Trial Combining Bevacizumab and Temsirolimus
    ANTICANCER RESEARCH 34: 1939-1946 (2014) Pediatric Patients with Refractory Central Nervous System Tumors: Experiences of a Clinical Trial Combining Bevacizumab and Temsirolimus SARINA A. PIHA-PAUL1*, SANG JOON SHIN1*, TRIBHAWAN VATS2, NANDITA GUHA-THAKURTA3, JOANN AARON1, MICHAEL RYTTING2, EUGENIE KLEINERMAN2 and RAZELLE KURZROCK4 Departments of 1Investigational Cancer Therapeutics (Phase I Clinical Trials Program), 2Pediatric Oncology, and 3Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, U.S.A.; 4Moores Cancer Center, The University of California San Diego, La Jolla, CA, U.S.A. Abstract. Background: Pre-clinical findings suggest that Primary central nervous system (CNS) malignancies are the combination treatment with bevacizumab and temsirolimus second most frequent tumors in pediatric patients, with 3.4 could be effective against malignant pediatric central cases per 100,000 person-years of the estimated incidence for nervous system (CNS) tumors. Patients and Methods: Six children and adolescents at or below 19 years of age in the pediatric patients were treated as part of a phase I trial with United States (1, 2). Advances in diagnostic and therapeutic intravenous temsirolimus 25 mg on days 1, 8, 15, and modalities have improved the 5-year survival rates of children bevacizumab at 5, 10, or 15 mg/kg on day 1 of each 21-day with primary CNS tumors (3). However, due to the cycle until disease progression or patient withdrawal. emergence of drug-resistant clones, the outcome is poor in Results: The median patient age was six years (range=3-14 progressive disease. During the past decade, targeted years). The primary diagnoses were glioblastoma multiforme therapies have been increasingly identified for the most (n=2), medullobalstoma (n=2), pontine glioma (n=1) and common adult malignancies.
    [Show full text]
  • WHO Drug Information Vol 22, No
    WHO Drug Information Vol 22, No. 1, 2008 World Health Organization WHO Drug Information Contents Challenges in Biotherapeutics Miglustat: withdrawal by manufacturer 21 Regulatory pathways for biosimilar Voluntary withdrawal of clobutinol cough products 3 syrup 22 Pharmacovigilance Focus Current Topics WHO Programme for International Drug Proposed harmonized requirements: Monitoring: annual meeting 6 licensing vaccines in the Americas 23 Sixteen types of counterfeit artesunate Safety and Efficacy Issues circulating in South-east Asia 24 Eastern Mediterranean Ministers tackle Recall of heparin products extended 10 high medicines prices 24 Contaminated heparin products recalled 10 DacartTM development terminated and LapdapTM recalled 11 ATC/DDD Classification Varenicline and suicide attempts 11 ATC/DDD Classification (temporary) 26 Norelgestromin-ethynil estradiol: infarction ATC/DDD Classification (final) 28 and thromboembolism 12 Emerging cardiovascular concerns with Consultation Document rosiglitazone 12 Disclosure of transdermal patches 13 International Pharmacopoeia Statement on safety of HPV vaccine 13 Cycloserine 30 IVIG: myocardial infarction, stroke and Cycloserine capsules 33 thrombosis 14 Erythropoietins: lower haemoglobin levels 15 Recent Publications, Erythropoietin-stimulating agents 15 Pregabalin: hypersensitivity reactions 16 Information and Events Cefepime: increased mortality? 16 Assessing the quality of herbal medicines: Mycophenolic acid: pregnancy loss and contaminants and residues 36 congenital malformation 17 Launch
    [Show full text]
  • 07052020 MR ASCO20 Curtain Raiser
    Media Release New data at the ASCO20 Virtual Scientific Program reflects Roche’s commitment to accelerating progress in cancer care First clinical data from tiragolumab, Roche’s novel anti-TIGIT cancer immunotherapy, in combination with Tecentriq® (atezolizumab) in patients with PD-L1-positive metastatic non- small cell lung cancer (NSCLC) Updated overall survival data for Alecensa® (alectinib), in people living with anaplastic lymphoma kinase (ALK)-positive metastatic NSCLC Key highlights to be shared on Roche’s ASCO virtual newsroom, 29 May 2020, 08:00 CEST Basel, 7 May 2020 - Roche (SIX: RO, ROG; OTCQX: RHHBY) today announced that new data from clinical trials of 19 approved and investigational medicines across 21 cancer types, will be presented at the ASCO20 Virtual Scientific Program organised by the American Society of Clinical Oncology (ASCO), which will be held 29-31 May, 2020. A total of 120 abstracts that include a Roche medicine will be presented at this year's meeting. "At ASCO, we will present new data from many investigational and approved medicines across our broad oncology portfolio," said Levi Garraway, M.D., Ph.D., Roche's Chief Medical Officer and Head of Global Product Development. “These efforts exemplify our long-standing commitment to improving outcomes for people with cancer, even during these unprecedented times. By integrating our medicines and diagnostics together with advanced insights and novel platforms, Roche is uniquely positioned to deliver the healthcare solutions of the future." Together with its partners, Roche is pioneering a comprehensive approach to cancer care, combining new diagnostics and treatments with innovative, integrated data and access solutions for approved medicines that will both personalise and transform the outcomes of people affected by this deadly disease.
    [Show full text]
  • MET Or NRAS Amplification Is an Acquired Resistance Mechanism to the Third-Generation EGFR Inhibitor Naquotinib
    www.nature.com/scientificreports OPEN MET or NRAS amplifcation is an acquired resistance mechanism to the third-generation EGFR inhibitor Received: 5 October 2017 Accepted: 16 January 2018 naquotinib Published: xx xx xxxx Kiichiro Ninomiya1, Kadoaki Ohashi1,2, Go Makimoto1, Shuta Tomida3, Hisao Higo1, Hiroe Kayatani1, Takashi Ninomiya1, Toshio Kubo4, Eiki Ichihara2, Katsuyuki Hotta5, Masahiro Tabata4, Yoshinobu Maeda1 & Katsuyuki Kiura2 As a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), osimeritnib is the standard treatment for patients with non-small cell lung cancer harboring the EGFR T790M mutation; however, acquired resistance inevitably develops. Therefore, a next-generation treatment strategy is warranted in the osimertinib era. We investigated the mechanism of resistance to a novel EGFR-TKI, naquotinib, with the goal of developing a novel treatment strategy. We established multiple naquotinib-resistant cell lines or osimertinib-resistant cells, two of which were derived from EGFR-TKI-naïve cells; the others were derived from geftinib- or afatinib-resistant cells harboring EGFR T790M. We comprehensively analyzed the RNA kinome sequence, but no universal gene alterations were detected in naquotinib-resistant cells. Neuroblastoma RAS viral oncogene homolog (NRAS) amplifcation was detected in naquotinib-resistant cells derived from geftinib-resistant cells. The combination therapy of MEK inhibitors and naquotinib exhibited a highly benefcial efect in resistant cells with NRAS amplifcation, but the combination of MEK inhibitors and osimertinib had limited efects on naquotinib-resistant cells. Moreover, the combination of MEK inhibitors and naquotinib inhibited the growth of osimertinib-resistant cells, while the combination of MEK inhibitors and osimertinib had little efect on osimertinib-resistant cells.
    [Show full text]
  • Haematological Malignancies
    Parikh_EU Haematology 03/03/2010 12:51 Page 43 Haematological Malignancies Clinical Management of Relapsed/Refractory Acute Myeloid Leukaemia Sameer A Parikh1 and Stefan Faderl2 1. Fellow; 2. Associate Professor of Medicine, Department of Leukemia, University of Texas MD Anderson Cancer Center DOI: 10.17925/EOH.2010.04.0.43 Abstract Conventional chemotherapy for patients with relapsed/refractory acute myeloid leukaemia (AML) remains unsatisfactory, with median survival ranging from 18 weeks following first relapse to six weeks in those with a second or higher relapse. The only potentially curative therapy is stem cell transplantation. Duration of first complete remission (CR) is the best predictor of response to salvage therapy. Novel therapies directed against a number of molecular aberrations associated with AML are being developed, including anti-CD33 monoclonal antibodies, FMS-like tyrosine kinase (FLT3) inhibitors, nucleoside analogues, hypomethylating agents and histone deacetylatase inhibitors, among others. Clinical trials combining novel agents with conventional chemotherapy are of particular interest, and definition of dose, schedule and combination partners remains an area of intense research. Keywords Acute myeloid leukaemia (AML), salvage therapy, chemotherapy, targeted therapy, nucleoside analogues, FLT3 inhibition, epigenetic therapy Disclosure: Sameer A Parikh has no conflicts of interst to declare. Stefan Faderl receives research funding from Genzyme Oncology and has been a member of advisory board meetings conducted by Genzyme Oncology. He is also a member of the speaker’s bureau of Eisai Inc. Received: 22 September 2009 Accepted: 23 February 2010 Citation: European Haematology, 2010;4:43–6 Correspondence: Stefan Faderl, Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, US.
    [Show full text]
  • DNA Replication During Acute MEK Inhibition Drives Acquisition of Resistance Through Amplification of the BRAF Oncogene
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.23.436572; this version posted March 23, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Acquisition of MEKi resistance during DNA replication in drug Channathodiyil et al. DNA replication during acute MEK inhibition drives acquisition of resistance through amplification of the BRAF oncogene Prasanna Channathodiyil1,2, Anne Segonds-Pichon3, Paul D. Smith4, Simon J. Cook5 and Jonathan Houseley1,6,* 1 Epigenetics Programme, Babraham Institute, Cambridge, UK 2 ORCID: 0000-0002-9381-6089 3 Babraham Bioinformatics, Babraham Institute, Cambridge, UK. ORCID: 0000-0002-8369- 4882 4 Oncology R&D, AstraZeneca CRUK Cambridge Institute, Cambridge, UK 5 Signalling programme, Babraham Institute, Cambridge, UK. ORCID: 0000-0001-9087-1616 6 ORCID: 0000-0001-8509-1500 * Corresponding author: [email protected] Abstract Mutations and gene amplifications that confer drug resistance emerge frequently during chemotherapy, but their mechanism and timing is poorly understood. Here, we investigate BRAFV600E amplification events that underlie resistance to the MEK inhibitor selumetinib (AZD6244/ARRY-142886) in COLO205 cells. We find that de novo focal BRAF amplification is the primary path to resistance irrespective of pre-existing amplifications. Although selumetinib causes long-term G1 arrest, we observe that cells stochastically re-enter the cell cycle during treatment without reactivation of ERK1/2 or induction of a normal proliferative gene expression programme. Genes encoding DNA replication and repair factors are downregulated during G1 arrest, but many are transiently induced when cells escape arrest and enter S and G2.
    [Show full text]
  • Monovalent Antibody Design and Mechanism of Action of Onartuzumab, a MET Antagonist with Anti-Tumor Activity As a Therapeutic Ag
    Monovalent antibody design and mechanism of PNAS PLUS action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent Mark Merchanta,1,2, Xiaolei Mab,2,3, Henry R. Maunc,2, Zhong Zhenga,2,4, Jing Penga, Mally Romeroa,5, Arthur Huangd,6, Nai-ying Yanga, Merry Nishimuraa, Joan Grevee, Lydia Santellc, Yu-Wen Zhangf, Yanli Suf, Dafna W. Kaufmanf, Karen L. Billecig, Elaine Maih, Barbara Moffatg,7, Amy Limi, Eileen T. Duenasi, Heidi S. Phillipsa, Hong Xiangj, Judy C. Youngh, George F. Vande Woudef, Mark S. Dennisd, Dorothea E. Reillyk, Ralph H. Schwalla,8, Melissa A. Starovasnikb, Robert A. Lazarusc, and Daniel G. Yansurad Departments of aTranslational Oncology, bStructural Biology, cEarly d e Discovery Biochemistry, Antibody Engineering, Biomedical Imaging, fi gProtein Chemistry, hBiochemical and Cellular Pharmacology, iPurification Signi cance Development, jPharmacokinetic and Pharmacodynamic Sciences, and kEarly Stage Cell Culture, Genentech, Inc., South San Francisco, CA 94080; and Therapeutic antibodies have revolutionized the treatment of hu- fLaboratory of Molecular Oncology, Van Andel Research Institute, Grand Rapids, MI 49503 man disease. Despite these advances, antibody bivalency limits their utility against some targets. Here, we describe the de- Edited by Richard A. Lerner, The Scripps Research Institute, La Jolla, CA, and velopment of a one-armed (monovalent) antibody, onartuzumab, approved June 3, 2013 (received for review February 15, 2013) targeting the receptor tyrosine kinase MET. While initial screening Binding of hepatocyte growth factor (HGF) to the receptor tyrosine of bivalent antibodies produced agonists of MET, engineering kinase MET is implicated in the malignant process of multiple can- them into monovalent antibodies produced antagonists instead.
    [Show full text]
  • Dual Targeting of HER2-Positive Cancer with Trastuzumab Emtansine and Pertuzumab: Critical Role for Neuregulin Blockade in Antitumor Response to Combination Therapy
    Published OnlineFirst October 4, 2013; DOI: 10.1158/1078-0432.CCR-13-0358 Clinical Cancer Cancer Therapy: Clinical Research See related article by Gwin and Spector, p. 278 Dual Targeting of HER2-Positive Cancer with Trastuzumab Emtansine and Pertuzumab: Critical Role for Neuregulin Blockade in Antitumor Response to Combination Therapy Gail D. Lewis Phillips1, Carter T. Fields1, Guangmin Li1, Donald Dowbenko1, Gabriele Schaefer1, Kathy Miller5, Fabrice Andre6, Howard A. Burris III8, Kathy S. Albain9, Nadia Harbeck10, Veronique Dieras7, Diana Crivellari11, Liang Fang2, Ellie Guardino3, Steven R. Olsen3, Lisa M. Crocker4, and Mark X. Sliwkowski1 Abstract Purpose: Targeting HER2 with multiple HER2-directed therapies represents a promising area of treatment for HER2-positive cancers. We investigated combining the HER2-directed antibody–drug con- jugate trastuzumab emtansine (T-DM1) with the HER2 dimerization inhibitor pertuzumab (Perjeta). Experimental Design: Drug combination studies with T-DM1 and pertuzumab were performed on cultured tumor cells and in mouse xenograft models of HER2-amplified cancer. In patients with HER2- positive locally advanced or metastatic breast cancer (mBC), T-DM1 was dose-escalated with a fixed standard pertuzumab dose in a 3þ3 phase Ib/II study design. Results: Treatment of HER2-overexpressing tumor cells in vitro with T-DM1 plus pertuzumab resulted in synergistic inhibition of cell proliferation and induction of apoptotic cell death. The presence of the HER3 ligand, heregulin (NRG-1b), reduced the cytotoxic activity of T-DM1 in a subset of breast cancer lines; this effect was reversed by the addition of pertuzumab. Results from mouse xenograft models showed enhanced antitumor efficacy with T-DM1 and pertuzumab resulting from the unique antitumor activities of each agent.
    [Show full text]