Correlation Between Gene Expression of IGF-1R Pathway Markers and Cetuximab Benefit in Metastatic Colorectal Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Correlation Between Gene Expression of IGF-1R Pathway Markers and Cetuximab Benefit in Metastatic Colorectal Cancer Published OnlineFirst January 31, 2012; DOI: 10.1158/1078-0432.CCR-11-1135 Clinical Cancer Predictive Biomarkers and Personalized Medicine Research Correlation between Gene Expression of IGF-1R Pathway Markers and Cetuximab Benefit in Metastatic Colorectal Cancer Fei Huang, Li-an Xu, and Shirin Khambata-Ford Abstract Purpose: This study examined potential correlations between markers related to the insulin-like growth factor-1 receptor (IGF-1R) pathway and clinical benefit from the anti–epidermal growth factor receptor (EGFR) monoclonal antibody cetuximab in metastatic colorectal cancer (mCRC). Experimental Design: Gene expression profiles for 70 pretreatment specimens from metastatic lesions of patients with chemorefractory mCRC receiving cetuximab monotherapy were analyzed using 74 predefined Gene-Chip probesets representing 33 unique IGF-1R pathway markers to determine correlations with progression-free survival (PFS) and disease control rate. Results: Higher IGF-1R, higher GRB7, and lower INSIG2 expression were associated with longer PFS with cetuximab in univariate analyses, particularly in patients with wild-type K-Ras tumors: median, 122 versus 60 days (P ¼ 0.01), 122 versus 57 days (P ¼ 0.011), and 57 versus 156 days (P < 0.0001), favoring higher IGF- 1R, higher GRB7, and lower INSIG2 expression, respectively. Lower IGF-1 expression was associated with a PFS benefit with cetuximab, whereas lower IGFBP3 and INSR expression levels showed trends for a PFS benefit. Lower INSIG2 expression (vs. higher expression) was associated with greater PFS in the high epiregulin-expressing group (P ¼ 0.001), but not in the low-expressing cohort suggesting an effect independent from the previously reported effect of epiregulin expression. Lower INSIG2 expression was also associated with higher disease control rate in the overall population (51.4% vs. 11.4%; P ¼ 0.001) and wild-type K-Ras subset (76.2% vs. 18.2%; P < 0.0001). Conclusions: These results suggest that markers of the IGF-1R pathway may play a role in predicting benefit from cetuximab therapy in mCRC. Additional clinical studies are warranted to validate these findings. Clin Cancer Res; 18(4); 1156–66. Ó2012 AACR. Introduction that the presence of K-Ras exon-2 mutations (codons 12 and 13) predicts lack of cetuximab benefit in mCRC, both as Cetuximab is an anti–epidermal growth factor receptor single-agent therapy in patients who are refractory to che- (EGFR) monoclonal antibody that has antitumor efficacy in motherapy and in combination with chemotherapy in metastatic colorectal cancer (mCRC; refs. 1, 2); locoregion- treatment-na€ve patients (2, 6–8). Whereas these original ally advanced recurrent or metastatic squamous cell carci- studies bundled all known exon-2 mutations as one single noma of the head and neck (3, 4); and advanced non–small "resistance genotype," more recent studies have indicated cell lung cancer (NSCLC; ref. 5). In unselected patients with that patients with tumors carrying the G13D mutation may mCRC, cetuximab significantly improved survival as a sin- benefit from therapy, suggesting that this mutation may not gle agent compared with best supportive care in patients have the same effect as the rest on exon-2 (9, 10). These previously treated with fluoropyrimidine, irinotecan, and findings represent a significant breakthrough in mCRC oxaliplatin (1), and when added to first-line FOLFIRI com- treatment because they allow cetuximab to be directed to pared with FOLFIRI alone (6). Multiple studies have shown patients who are most likely to benefit. However, not all patients with wild-type K-Ras benefit from cetuximab-based therapy, and further refinement of patient selection is Authors' Affiliation: Bristol-Myers Squibb Co., Princeton, New Jersey desirable. Note: Supplementary data for this article are available at Clinical Cancer In chemotherapy (particularly irinotecan)-refractory Research Online (http://clincancerres.aacrjournals.org/). patients, there is a growing list of candidate predictive Corresponding Author: Fei Huang, Bristol-Myers Squibb Co., Route 206 markers under investigation. These include EGFR gene copy and Province Line Rd., Room E1.293, Princeton, NJ 08453. Phone: 609- number measured by FISH, ligand gene expression (amphir- 252-4891; Fax: 1-609-818-5839; E-mail: [email protected] egulin and epiregulin [EREG]) or polymorphisms (EGF), doi: 10.1158/1078-0432.CCR-11-1135 mutations in components of the MAP-Ras-Raf pathway, Ó2012 American Association for Cancer Research. loss of the PTEN gene, and PIK3CA expression levels or 1156 Clin Cancer Res; 18(4) February 15, 2012 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2012 American Association for Cancer Research. Published OnlineFirst January 31, 2012; DOI: 10.1158/1078-0432.CCR-11-1135 IGF-1R Pathway Markers and Cetuximab Benefit in CRC activated protein kinase (MAPK) and phosphoinositide Translational Relevance 3-kinase (PI3K), and may work collaterally to drive tumor growth and survival (reviewed in ref. 30). The IGF-1R Patients with metastatic colorectal cancer (mCRC) pathway appears to regulate EGFR signaling and, recipro- whose tumors harbor the wild-type K-Ras gene may cally, EGFR ligands may promote tumor survival through derive benefit from treatment with the anti–epidermal IGF-1R (31–33). Indeed, overexpression of the EGFR ligand growth factor receptor (EGFR) monoclonal antibody amphiregulin in NSCLC cell lines stimulates IGF-1R, lead- cetuximab, whereas those patients with tumors bearing ing to IGF-1 and amphiregulin secretion and inhibition of certain K-Ras mutations do not benefit. However, not all apoptosis (32). EGFR tyrosine kinase inhibitors (TKI) can patients with wild-type K-Ras tumors benefit from cetux- induce EGFR/IGF-1R heterodimerization and alter the imab, underscoring the need for additional markers to interactions between IGF-1R and insulin receptor (INSR) help in patient selection. Preclinical evidence suggests substrate (IRS)-1 in cancer cells, activating downstream that the EGFR and insulin-like growth factor-1 receptor signaling and resulting in resistance to further TKI treatment (IGF-1R) pathways interact to drive tumor growth and (34–36). Conversely, coinhibition of both pathways results survival. This study evaluated whether expression levels in synergistic antitumor effects in several preclinical cancer of the IGF-1R pathway markers correlate with cetuximab models (36–41). benefit in patients with chemorefractory mCRC, includ- Therefore, it is reasonable to hypothesize that molecular ing the subset of patients with wild-type K-Ras. The markers associated with the IGF-1R pathway may affect the results provide initial evidence that markers of the antitumor activity of EGFR inhibitors. In this study, we IGF-1R pathway may predict benefit from cetuximab analyzed the gene expression levels of IGF-1R pathway therapy in mCRC, but this preliminary finding requires components previously identified as potential points of validation in additional clinical studies. EGFR-IGF-1R cross-talk (15) and investigated their poten- tial correlation with cetuximab benefit in patients with chemorefractory mCRC, both in the overall population and in the group with K-Ras wild-type tumors. mutations. All have been reported to affect the clinical activity of cetuximab (11–25), mostly in relatively small Methods patient cohorts, often without randomized controls. Com- bining the testing for K-Ras mutation with one or more of Patients and study design these markers could further enhance patient selection and The CA225-045 trial was a phase II pharmacogenomic the overall therapeutic index of cetuximab in mCRC. The study in which 110 patients with chemorefractory mCRC advantages of this approach have been illustrated by the received single-agent cetuximab after failing at least one enrichment in responding patients achieved with nested chemotherapeutic regimen for advanced disease or refusing mutational screening of tumors for K-Ras, B-Raf, N-Ras, prior treatment. Details of the study design and patient PIK3CA (26), or with the combimarker of K-Ras mutation cohort have been described previously (13). Patients plus EREG expression (16). received cetuximab at its standard dose (400 mg/m2 loading In previously untreated patients with mCRC receiving dose; then 250 mg/m2 weekly) for 3 weeks and then were first-line therapy, however, data from randomized con- eligible for dose-escalation every 3 weeks to a maximum of trolled studies are lacking for most of the candidate predic- 400 mg/m2 weekly, provided they had not experienced tive markers for anti-EGFR therapy. One in particular, B-Raf greater than grade II skin rashes. Tumor responses were V600E mutation, has not upheld the potentially predictive evaluated every 9 weeks according to modified WHO cri- value consistently reported in noncontrolled retrospective teria (42). The disease control rate (DCR) was defined as the series of irinotecan-refractory patients (23, 25, 26), showing proportion of patients with a complete response (CR), instead a notable prognostic effect in controlled studies partial response (PR), or stable disease (SD). Progression- of untreated patients, regardless of cetuximab treatment free survival (PFS) was defined as the time from enrollment (6, 27). The data are therefore conflicting and preclude firm until disease progression or death. The study protocol was practical recommendations for any predictive marker other approved by the Institutional Review Board at each partic- than K-Ras exon-2 mutations. Whether the apparent
Recommended publications
  • Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients
    antibodies Review Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients Andrew T. Lucas 1,2,3,*, Ryan Robinson 3, Allison N. Schorzman 2, Joseph A. Piscitelli 1, Juan F. Razo 1 and William C. Zamboni 1,2,3 1 University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; [email protected] (J.A.P.); [email protected] (J.F.R.); [email protected] (W.C.Z.) 2 Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; [email protected] 3 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-919-966-5242; Fax: +1-919-966-5863 Received: 30 November 2018; Accepted: 22 December 2018; Published: 1 January 2019 Abstract: The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution.
    [Show full text]
  • Etude Des Résistances Adaptatives Aux Inhibiteurs De Tyrosine Kinase De L’EGFR Dans Les Cancers Bronchiques
    Thèse d’exercice Faculté de Pharmacie Année 2020 Thèse N° MÉMOIRE DU DIPLÔME D'ÉTUDES SPÉCIALISÉES D’INNOVATION PHARMACEUTIQUE ET RECHERCHE TENANT LIEU DE THÈSE D’EXERCICE POUR LE DIPLÔME D’ÉTAT DE DOCTEUR ENPHARMACIE Présentée et soutenue publiquement le 24 septembre 2020 par Sarah FIGAROL Née le 19 septembre 1989 à Toulouse Etude des résistances adaptatives aux inhibiteurs de tyrosine kinase de l’EGFR dans les cancers bronchiques Thèse dirigée par Gilles Favre Examinateurs : M. Franck Saint-Marcoux, président du jury M.Gilles Favre, directeur de thèse M. Jean-Marie Canonge, juge M. Julien Mazières, juge M. Antonio Maraver, juge Thèse d’exercice Faculté de Pharmacie Année 2020 Thèse N° MÉMOIRE DU DIPLÔME D'ÉTUDES SPÉCIALISÉES D’INNOVATION PHARMACEUTIQUE ET RECHERCHE TENANT LIEU DE THÈSE D’EXERCICE POUR LE DIPLÔME D’ÉTAT DE DOCTEUR ENPHARMACIE Présentée et soutenue publiquement le 24 septembre 2020 par Sarah FIGAROL Née le 19 septembre 1989 à Toulouse Etude des résistances adaptatives aux inhibiteurs de tyrosine kinase de l’EGFR dans les cancers bronchiques Thèse dirigée par Gilles Favre Examinateurs : M. Franck Saint-Marcoux, président du jury M.Gilles Favre, directeur de thèse M. Jean-Marie Canonge, juge M. Julien Mazières, juge M. Antonio Maraver, juge Sarah FIGAROL | Thèse d’exercice | Université de Limoges |2020 3 Licence CC BY-NC-ND 3.0 Liste des enseignants Le 1er septembre 2019 PROFESSEURS : BATTU Serge CHIMIE ANALYTIQUE CARDOT Philippe CHIMIE ANALYTIQUE ET BROMATOLOGIE DESMOULIERE Alexis PHYSIOLOGIE DUROUX Jean-Luc BIOPHYSIQUE,
    [Show full text]
  • The Role of Biological Therapy in Metastatic Colorectal Cancer After First-Line Treatment: a Meta-Analysis of Randomised Trials
    REVIEW British Journal of Cancer (2014) 111, 1122–1131 | doi: 10.1038/bjc.2014.404 Keywords: colorectal; biological; meta-analysis The role of biological therapy in metastatic colorectal cancer after first-line treatment: a meta-analysis of randomised trials E Segelov1, D Chan*,2, J Shapiro3, T J Price4, C S Karapetis5, N C Tebbutt6 and N Pavlakis2 1St Vincent’s Clinical School, University of New South Wales, Sydney, NSW 2052, Australia; 2Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia; 3Monash University and Cabrini Hospital, Melbourne, VIC 3800, Australia; 4The Queen Elizabeth Hospital and University of Adelaide, Woodville South, SA 5011, Australia; 5Flinders University and Flinders Medical Centre, Flinders Centre for Innovation in Cancer, Bedford Park, SA, 5042, Australia and 6Austin Health, VIC 3084, Australia Purpose: Biologic agents have achieved variable results in relapsed metastatic colorectal cancer (mCRC). Systematic meta-analysis was undertaken to determine the efficacy of biological therapy. Methods: Major databases were searched for randomised studies of mCRC after first-line treatment comparing (1) standard treatment plus biologic agent with standard treatment or (2) standard treatment with biologic agent with the same treatment with different biologic agent(s). Data were extracted on study design, participants, interventions and outcomes. Study quality was assessed using the MERGE criteria. Comparable data were pooled for meta-analysis. Results: Twenty eligible studies with 8225 patients were identified. The use of any biologic therapy improved overall survival with hazard ratio (HR) 0.87 (95% confidence interval (CI) 0.82–0.91, Po0.00001), progression-free survival (PFS) with HR 0.71 (95% CI 0.67–0.74, Po0.0001) and overall response rate (ORR) with odds ratio (OR) 2.38 (95% CI 2.03–2.78, Po0.00001).
    [Show full text]
  • Targeted and Novel Therapy in Advanced Gastric Cancer Julie H
    Selim et al. Exp Hematol Oncol (2019) 8:25 https://doi.org/10.1186/s40164-019-0149-6 Experimental Hematology & Oncology REVIEW Open Access Targeted and novel therapy in advanced gastric cancer Julie H. Selim1 , Shagufta Shaheen2 , Wei‑Chun Sheu3 and Chung‑Tsen Hsueh4* Abstract The systemic treatment options for advanced gastric cancer (GC) have evolved rapidly in recent years. We have reviewed the recent data of clinical trial incorporating targeted agents, including inhibitors of angiogenesis, human epidermal growth factor receptor 2 (HER2), mesenchymal–epithelial transition, epidermal growth factor receptor, mammalian target of rapamycin, claudin‑18.2, programmed death‑1 and DNA. Addition of trastuzumab to platinum‑ based chemotherapy has become standard of care as front‑line therapy in advanced GC overexpressing HER2. In the second‑line setting, ramucirumab with paclitaxel signifcantly improves overall survival compared to paclitaxel alone. For patients with refractory disease, apatinib, nivolumab, ramucirumab and TAS‑102 have demonstrated single‑agent activity with improved overall survival compared to placebo alone. Pembrolizumab has demonstrated more than 50% response rate in microsatellite instability‑high tumors, 15% response rate in tumors expressing programmed death ligand 1, and non‑inferior outcome in frst‑line treatment compared to chemotherapy. This review summarizes the current state and progress of research on targeted therapy for advanced GC. Keywords: Gastric cancer, Targeted therapy, Human epidermal growth factor receptor 2, Programmed death‑1, Vascular endothelial growth factor receptor 2 Background GC mortality which is consistent with overall decrease in Gastric cancer (GC), including adenocarcinoma of the GC-related deaths [4]. gastroesophageal junction (GEJ) and stomach, is the ffth Tere have been several eforts to perform large-scale most common cancer and the third leading cause of can- molecular profling and classifcation of GC.
    [Show full text]
  • IGF System in Sarcomas: a Crucial Pathway with Many Unknowns to Exploit for Therapy
    61 1 Journal of Molecular C Mancarella and Targeting IGF system in sarcoma 61:1 T45–T60 Endocrinology K Scotlandi THEMATIC REVIEW 40 YEARS OF IGF1 IGF system in sarcomas: a crucial pathway with many unknowns to exploit for therapy Caterina Mancarella and Katia Scotlandi Experimental Oncology Lab, CRS Development of Biomolecular Therapies, Orthopaedic Rizzoli Institute, Bologna, Italy Correspondence should be addressed to K Scotlandi: [email protected] This paper forms part of a special section on 40 Years of IGF1. The guest editors for this section were Derek LeRoith and Emily Gallagher. Abstract The insulin-like growth factor (IGF) system has gained substantial interest due to its Key Words involvement in regulating cell proliferation, differentiation and survival during anoikis f sarcomas and after conventional and targeted therapies. However, results from clinical trials have f IGF system been largely disappointing, with only a few but notable exceptions, such as trials targeting f targeted therapy sarcomas, especially Ewing sarcoma. This review highlights key studies focusing on IGF f clinical trials signaling in sarcomas, specifically studies underscoring the properties that make this system an attractive therapeutic target and identifies new relationships that may be exploited. This review discusses the potential roles of IGF2 mRNA-binding proteins (IGF2BPs), discoidin domain receptors (DDRs) and metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) in regulating the IGF system. Deeper investigation of these novel regulators of the IGF system may help us to further elucidate the spatial and temporal control of the IGF Journal of Molecular axis, as understanding the control of this axis is essential for future clinical studies.
    [Show full text]
  • Therapeutic Targeting of the IGF Axis
    cells Review Therapeutic Targeting of the IGF Axis Eliot Osher and Valentine M. Macaulay * Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK * Correspondence: [email protected]; Tel.: +44-1865617337 Received: 8 July 2019; Accepted: 9 August 2019; Published: 14 August 2019 Abstract: The insulin like growth factor (IGF) axis plays a fundamental role in normal growth and development, and when deregulated makes an important contribution to disease. Here, we review the functions mediated by ligand-induced IGF axis activation, and discuss the evidence for the involvement of IGF signaling in the pathogenesis of cancer, endocrine disorders including acromegaly, diabetes and thyroid eye disease, skin diseases such as acne and psoriasis, and the frailty that accompanies aging. We discuss the use of IGF axis inhibitors, focusing on the different approaches that have been taken to develop effective and tolerable ways to block this important signaling pathway. We outline the advantages and disadvantages of each approach, and discuss progress in evaluating these agents, including factors that contributed to the failure of many of these novel therapeutics in early phase cancer trials. Finally, we summarize grounds for cautious optimism for ongoing and future studies of IGF blockade in cancer and non-malignant disorders including thyroid eye disease and aging. Keywords: IGF; type 1 IGF receptor; IGF-1R; cancer; acromegaly; ophthalmopathy; IGF inhibitor 1. Introduction Insulin like growth factors (IGFs) are small (~7.5 kDa) ligands that play a critical role in many biological processes including proliferation and protection from apoptosis and normal somatic growth and development [1]. IGFs are members of a ligand family that includes insulin, a dipeptide comprised of A and B chains linked via two disulfide bonds, with a third disulfide linkage within the A chain.
    [Show full text]
  • Primary and Acquired Resistance to Immunotherapy in Lung Cancer: Unveiling the Mechanisms Underlying of Immune Checkpoint Blockade Therapy
    cancers Review Primary and Acquired Resistance to Immunotherapy in Lung Cancer: Unveiling the Mechanisms Underlying of Immune Checkpoint Blockade Therapy Laura Boyero 1 , Amparo Sánchez-Gastaldo 2, Miriam Alonso 2, 1 1,2,3, , 1,2, , José Francisco Noguera-Uclés , Sonia Molina-Pinelo * y and Reyes Bernabé-Caro * y 1 Institute of Biomedicine of Seville (IBiS) (HUVR, CSIC, Universidad de Sevilla), 41013 Seville, Spain; [email protected] (L.B.); [email protected] (J.F.N.-U.) 2 Medical Oncology Department, Hospital Universitario Virgen del Rocio, 41013 Seville, Spain; [email protected] (A.S.-G.); [email protected] (M.A.) 3 Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain * Correspondence: [email protected] (S.M.-P.); [email protected] (R.B.-C.) These authors contributed equally to this work. y Received: 16 November 2020; Accepted: 9 December 2020; Published: 11 December 2020 Simple Summary: Immuno-oncology has redefined the treatment of lung cancer, with the ultimate goal being the reactivation of the anti-tumor immune response. This has led to the development of several therapeutic strategies focused in this direction. However, a high percentage of lung cancer patients do not respond to these therapies or their responses are transient. Here, we summarized the impact of immunotherapy on lung cancer patients in the latest clinical trials conducted on this disease. As well as the mechanisms of primary and acquired resistance to immunotherapy in this disease. Abstract: After several decades without maintained responses or long-term survival of patients with lung cancer, novel therapies have emerged as a hopeful milestone in this research field.
    [Show full text]
  • Anti-EGFR Treatment for EGFR-Amplified Gastroesophageal Adenocarcinoma
    Published OnlineFirst February 15, 2018; DOI: 10.1158/2159-8290.CD-17-1260 RESEARCH ARTICLE Targeted Therapies for Targeted Populations: Anti-EGFR Treatment for EGFR -Amplifi ed Gastroesophageal Adenocarcinoma Steven B. Maron 1 , Lindsay Alpert 2 , Heewon A. Kwak 2 , Samantha Lomnicki 1 , Leah Chase 1 , David Xu 1 , Emily O’Day1 , Rebecca J. Nagy 3 , Richard B. Lanman 3 , Fabiola Cecchi 4 , Todd Hembrough 4 , Alexa Schrock 5 , John Hart2 , Shu-Yuan Xiao 2 , Namrata Setia 2 , and Daniel V.T. Catenacci 1 ABSTRACT Previous anti-EGFR trials in unselected patients with gastroesophageal adeno- carcinoma (GEA) were resoundingly negative. We identifi ed EGFR amplifi cation in 5% (19/363) of patients at the University of Chicago, including 6% (8/140) who were prospectively screened with intention-to-treat using anti-EGFR therapy. Seven patients received ≥1 dose of treat- ment: three fi rst-line FOLFOX plus ABT-806, one second-line FOLFIRI plus cetuximab, and three third/ fourth-line cetuximab alone. Treatment achieved objective response in 58% (4/7) and disease control in 100% (7/7) with a median progression-free survival of 10 months. Pretreatment and posttreatment tumor next-generation sequencing (NGS), serial plasma circulating tumor DNA (ctDNA) NGS, and tumor IHC/FISH for EGFR revealed preexisting and/or acquired genomic events, including EGFR- negative clones, PTEN deletion, KRAS amplifi cation/mutation,NRAS, MYC , and HER2 amplifi cation, andGNAS mutations serving as mechanisms of resistance. Two evaluable patients demonstrated interval increase of CD3 + infi ltrate, including one who demonstrated increased NKp46+ , and PD-L1 IHC expression from baseline, suggesting an immune therapeutic mechanism of action.
    [Show full text]
  • Progress in Pancreatic Ductal Adenocarcinoma (PDAC) Research Working Group (PDAC Progress Working Group)
    National Cancer Institute Clinical Trials and Translational Research Advisory Committee (CTAC) Progress in Pancreatic Ductal Adenocarcinoma (PDAC) Research Working Group (PDAC Progress Working Group) Working Group Report March 6, 2019 The report was accepted at the March 6, 2019 CTAC Meeting ______________________________________________________________________________________________________ PDAC Progress WG Report: Accepted by the Clinical Trials and Translational Research Advisory Committee on March 6, 2019 Page 1 TABLE OF CONTENTS Page National Cancer Institute Clinical Trials and Translational Research Advisory Committee (CTAC) Progress in Pancreatic Ductal Adenocarcinoma (PDAC) Research 3 Working Group (PDAC Progress Working Group) Report Appendix 1: Progress in Pancreatic Ductal Adenocarcinoma (PDAC) Research 16 Working Group (PDAC Progress WG) 2018 Roster Appendix 2: Funded Project Summary FY 2015 – FY 2017 • NIH PDAC Funded Projects, Initiative 1: Understanding the 18 Biological Relationship Between PDAC and Diabetes Mellitus • NIH PDAC Funded Projects, Initiative 2: Evaluating Longitudinal Screening Protocols, for Biomarkers for Earl Detection of PDAC 20 and its Precursors • NIH PDAC Funded Projects, Initiative 3: Study New Therapeutic 28 Strategies in Immunotherapy • NIH PDAC Funded Projects, Initiative 4: Developing New Treatment Approaches that Interfere with RAS Oncogene- 33 Dependent Signaling Pathways • NIH PDAC Funded Projects, No Related to the Framework t 38 Initiatives Appendix 3: PDAC Publications Supported by Grants
    [Show full text]
  • The Two Tontti Tudiul Lui Hi Ha Unit
    THETWO TONTTI USTUDIUL 20170267753A1 LUI HI HA UNIT ( 19) United States (12 ) Patent Application Publication (10 ) Pub. No. : US 2017 /0267753 A1 Ehrenpreis (43 ) Pub . Date : Sep . 21 , 2017 ( 54 ) COMBINATION THERAPY FOR (52 ) U .S . CI. CO - ADMINISTRATION OF MONOCLONAL CPC .. .. CO7K 16 / 241 ( 2013 .01 ) ; A61K 39 / 3955 ANTIBODIES ( 2013 .01 ) ; A61K 31 /4706 ( 2013 .01 ) ; A61K 31 / 165 ( 2013 .01 ) ; CO7K 2317 /21 (2013 . 01 ) ; (71 ) Applicant: Eli D Ehrenpreis , Skokie , IL (US ) CO7K 2317/ 24 ( 2013. 01 ) ; A61K 2039/ 505 ( 2013 .01 ) (72 ) Inventor : Eli D Ehrenpreis, Skokie , IL (US ) (57 ) ABSTRACT Disclosed are methods for enhancing the efficacy of mono (21 ) Appl. No. : 15 /605 ,212 clonal antibody therapy , which entails co - administering a therapeutic monoclonal antibody , or a functional fragment (22 ) Filed : May 25 , 2017 thereof, and an effective amount of colchicine or hydroxy chloroquine , or a combination thereof, to a patient in need Related U . S . Application Data thereof . Also disclosed are methods of prolonging or increasing the time a monoclonal antibody remains in the (63 ) Continuation - in - part of application No . 14 / 947 , 193 , circulation of a patient, which entails co - administering a filed on Nov. 20 , 2015 . therapeutic monoclonal antibody , or a functional fragment ( 60 ) Provisional application No . 62/ 082, 682 , filed on Nov . of the monoclonal antibody , and an effective amount of 21 , 2014 . colchicine or hydroxychloroquine , or a combination thereof, to a patient in need thereof, wherein the time themonoclonal antibody remains in the circulation ( e . g . , blood serum ) of the Publication Classification patient is increased relative to the same regimen of admin (51 ) Int .
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2017/0172932 A1 Peyman (43) Pub
    US 20170172932A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2017/0172932 A1 Peyman (43) Pub. Date: Jun. 22, 2017 (54) EARLY CANCER DETECTION AND A 6LX 39/395 (2006.01) ENHANCED IMMUNOTHERAPY A61R 4I/00 (2006.01) (52) U.S. Cl. (71) Applicant: Gholam A. Peyman, Sun City, AZ CPC .......... A61K 9/50 (2013.01); A61K 39/39558 (US) (2013.01); A61K 4I/0052 (2013.01); A61 K 48/00 (2013.01); A61K 35/17 (2013.01); A61 K (72) Inventor: sham A. Peyman, Sun City, AZ 35/15 (2013.01); A61K 2035/124 (2013.01) (21) Appl. No.: 15/143,981 (57) ABSTRACT (22) Filed: May 2, 2016 A method of therapy for a tumor or other pathology by administering a combination of thermotherapy and immu Related U.S. Application Data notherapy optionally combined with gene delivery. The combination therapy beneficially treats the tumor and pre (63) Continuation-in-part of application No. 14/976,321, vents tumor recurrence, either locally or at a different site, by filed on Dec. 21, 2015. boosting the patient’s immune response both at the time or original therapy and/or for later therapy. With respect to Publication Classification gene delivery, the inventive method may be used in cancer (51) Int. Cl. therapy, but is not limited to such use; it will be appreciated A 6LX 9/50 (2006.01) that the inventive method may be used for gene delivery in A6 IK 35/5 (2006.01) general. The controlled and precise application of thermal A6 IK 4.8/00 (2006.01) energy enhances gene transfer to any cell, whether the cell A 6LX 35/7 (2006.01) is a neoplastic cell, a pre-neoplastic cell, or a normal cell.
    [Show full text]
  • A Focus on Breast Cancer
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Pharmacol Manuscript Author Ther. Author Manuscript Author manuscript; available in PMC 2017 May 01. Published in final edited form as: Pharmacol Ther. 2016 May ; 161: 79–96. doi:10.1016/j.pharmthera.2016.03.003. Emerging therapeutic targets in metastatic progression: a focus on breast cancer Zhuo Li and Yibin Kang* Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States Abstract Metastasis is the underlying cause of death for the majority of breast cancer patients. Despite significant advances in recent years in basic research and clinical development, therapies that specifically target metastatic breast cancer remain inadequate, and represents the single greatest obstacle to reducing mortality of late-stage breast cancer. Recent efforts have leveraged genomic analysis of breast cancer and molecular dissection of tumor-stromal cross-talk to uncover a number of promising candidates for targeted treatment of metastatic breast cancer. Rational combinations of therapeutic agents targeting tumor-intrinsic properties and microenvironmental components provide a promising strategy to develop precision treatments with higher specificity and less toxicity. In this review, we discuss the emerging therapeutic targets in breast cancer metastasis, from tumor-intrinsic pathways to those that involve the host tissue components, including the immune system. Keywords Breast cancer; Metastasis; Targeted therapy; Tumor microenvironment; Immunotherapy 1. Introduction The overall 5-year survival rate for breast cancer currently stands at 90% — a dramatic improvement over the 63% survival rate in the early 1960s. When stratified by stage, the 5- year survival rates have increased to 99% for localized disease and 85% for regional advanced disease, a trend that can be attributed to early diagnoses and better treatment regimens.
    [Show full text]