1
|
Mellinghoff IK, Cloughesy TF. Balancing Risk and Efficiency in Drug Development for Rare and Challenging Tumors: A New Paradigm for Glioma. J Clin Oncol 2022; 40:3510-3519. [PMID: 35201903 PMCID: PMC10166355 DOI: 10.1200/jco.21.02166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 12/14/2022] Open
Abstract
The process of developing cancer therapies is well established and has enabled the incorporation of many new drugs and classes of agents into the standard of care for common cancers. Clinical drug development is fundamentally different for rare and difficult-to-treat solid tumors, such as glioma or pancreatic cancer. The failure to develop effective new agents for the latter diseases has discouraged the development of therapeutics for these cancers. Using glioma as an example, we describe a process toward obtaining more reliable early-stage signals of drug activity and a process toward translating those signals into clinical benefits with more efficient late-stage development. If linked together, these processes should increase the likelihood of benefit in late-stage settings at a lower cost and encourage more drug development for patients with rare and difficult-to-treat cancers.
Collapse
Affiliation(s)
- Ingo K. Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Timothy F. Cloughesy
- Department of Neurology, Ronald Reagan UCLA Medical Center, University of California, Los Angeles, CA
| |
Collapse
|
2
|
Affolter A, Lammert A, Kern J, Scherl C, Rotter N. Precision Medicine Gains Momentum: Novel 3D Models and Stem Cell-Based Approaches in Head and Neck Cancer. Front Cell Dev Biol 2021; 9:666515. [PMID: 34307351 PMCID: PMC8296983 DOI: 10.3389/fcell.2021.666515] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the current progress in the development of new concepts of precision medicine for head and neck squamous cell carcinoma (HNSCC), in particular targeted therapies and immune checkpoint inhibition (CPI), overall survival rates have not improved during the last decades. This is, on the one hand, caused by the fact that a significant number of patients presents with late stage disease at the time of diagnosis, on the other hand HNSCC frequently develop therapeutic resistance. Distinct intratumoral and intertumoral heterogeneity is one of the strongest features in HNSCC and has hindered both the identification of specific biomarkers and the establishment of targeted therapies for this disease so far. To date, there is a paucity of reliable preclinical models, particularly those that can predict responses to immune CPI, as these models require an intact tumor microenvironment (TME). The "ideal" preclinical cancer model is supposed to take both the TME as well as tumor heterogeneity into account. Although HNSCC patients are frequently studied in clinical trials, there is a lack of reliable prognostic biomarkers allowing a better stratification of individuals who might benefit from new concepts of targeted or immunotherapeutic strategies. Emerging evidence indicates that cancer stem cells (CSCs) are highly tumorigenic. Through the process of stemness, epithelial cells acquire an invasive phenotype contributing to metastasis and recurrence. Specific markers for CSC such as CD133 and CD44 expression and ALDH activity help to identify CSC in HNSCC. For the majority of patients, allocation of treatment regimens is simply based on histological diagnosis and on tumor location and disease staging (clinical risk assessments) rather than on specific or individual tumor biology. Hence there is an urgent need for tools to stratify HNSCC patients and pave the way for personalized therapeutic options. This work reviews the current literature on novel approaches in implementing three-dimensional (3D) HNSCC in vitro and in vivo tumor models in the clinical daily routine. Stem-cell based assays will be particularly discussed. Those models are highly anticipated to serve as a preclinical prediction platform for the evaluation of stable biomarkers and for therapeutic efficacy testing.
Collapse
Affiliation(s)
- Annette Affolter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | |
Collapse
|
3
|
Duska LR, Filiaci VL, Walker JL, Holman LL, Hill EK, Moore RG, Ring KL, Pearl ML, Muller CY, Kushnir CL, Lankes HA, Samuelson MI, Carrick KS, Rajan A, Rodgers WH, Kohn EC, Piekarz R, Leslie KK. A Surgical Window Trial Evaluating Medroxyprogesterone Acetate with or without Entinostat in Patients with Endometrial Cancer and Validation of Biomarkers of Cellular Response. Clin Cancer Res 2021; 27:2734-2741. [PMID: 33766814 DOI: 10.1158/1078-0432.ccr-20-4618] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/23/2021] [Accepted: 02/26/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE This surgical window of opportunity (window) study assessed the short-term effect of medroxyprogesterone acetate (MPA) alone versus MPA plus the histone deacetylase (HDAC) inhibitor entinostat on regulation of progesterone receptor (PR) in women with newly diagnosed endometrioid endometrial adenocarcinoma. PATIENTS AND METHODS This multisite, randomized, open-label surgical window study treated women intramuscularly on day 1 with 400 mg MPA. Entinostat given 5 mg by mouth on days 1, 8, and 15 was randomly assigned with equal probability. Surgery followed on days 21-24. Pretreatment and posttreatment tissue was assessed for PR H-scores, Ki-67 levels, and histologic response. RESULTS Fifty patients were accrued in 4 months; 22 and 20 participants had PR evaluable pretreatment and posttreatment slides in the MPA and MPA/entinostat arms, respectively. Median posttreatment PR H-scores were significantly lower than pretreatment H-scores in both arms but did not differ significantly (MPA: 247 vs. 27, MPA/entinostat 260 vs. 23, respectively, P = 0.87). Decreased Ki-67 was shown in 90% treated with MPA/entinostat compared with 68% treated with MPA alone (P = 0.13). Median PR H-score decreases were larger when Ki-67 was decreased (208) versus not decreased (45). The decrease in PR pretreatment versus posttreatment was associated with loss of Ki-67 nuclear staining, consistent with reduced cellular proliferation (P < 0.008). CONCLUSIONS This surgical window trial rapidly accrued in a multisite setting and evaluated PR as its primary endpoint and Ki-67 as secondary endpoint. Despite no immediate effect of entinostat on PR in this short-term study, lessons learned can inform future window and treatment trials.
Collapse
Affiliation(s)
- Linda R Duska
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Virginia School of Medicine, Charlottesville, Virginia.
| | - Virginia L Filiaci
- NRG Oncology SDMC, CTD Division, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Joan L Walker
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Laura L Holman
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Emily K Hill
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Richard G Moore
- Department of Obstetrics and Gynecology, Wilmont Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | - Kari L Ring
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Michael L Pearl
- Department of Obstetrics, Gynecology and Reproductive Medicine, Division of Gynecologic Oncology, Stony Brook University Medical Center, Stony Brook, New York
| | - Carolyn Y Muller
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of New Mexico, Albuquerque, New Mexico
| | - Christina L Kushnir
- Department of Obstetrics and Gynecology, Women's Cancer Center, Las Vegas, Nevada
| | - Heather A Lankes
- NRG Oncology, Operations Center-Philadelphia East, Philadelphia, Pennsylvania.,Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Megan I Samuelson
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Kelley S Carrick
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Anand Rajan
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - William H Rodgers
- Department of Pathology, New York Hospital Queens, Weill Medical College of Cornell University, Flushing, New York
| | - Elise C Kohn
- Cancer Therapy Evaluation Program, NCI, Rockville, Maryland
| | | | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW Window of Opportunity (WOO) studies have gain their place in current clinical and translational research in breast cancer patients. This review provides current information and future applications of this specific type of research. RECENT FINDINGS So far, WOO trials in breast cancer patients have demonstrated their utility in breast cancer research as: first they allow administering a treatment for a short period of time to treatment-naïve patients whose tumors have not developed mechanisms of resistance or heterogeneity because of previous therapies. Second, it brings a unique opportunity for translational research providing easy access to tumor tissue in order to evaluate antitumor effect from initial biopsy and from surgical resection specimen. They provide the perfect scenario for biomarker discovery and validation in an efficient and timely manner and valuable information about drug pharmacodynamics. Several issues need to be contemplated when designing and performing this type of trials including choice of a biological surrogate endpoint of efficacy as standard clinical activity endpoints are not feasible. SUMMARY Despite some limitations like the absence of information about secondary mechanisms of resistance, WOO trials represent an important support for drug development and biomarker discovery in breast cancer patients.
Collapse
|
5
|
Robertson JFR, Coleman RE, Cheung KL, Evans A, Holcombe C, Skene A, Rea D, Ahmed S, Jahan A, Horgan K, Rauchhaus P, Littleford R, Cheung SYA, Cullberg M, de Bruin EC, Koulai L, Lindemann JPO, Pass M, Rugman P, Schiavon G, Deb R, Finlay P, Foxley A, Gee JMW. Proliferation and AKT Activity Biomarker Analyses after Capivasertib (AZD5363) Treatment of Patients with ER + Invasive Breast Cancer (STAKT). Clin Cancer Res 2020; 26:1574-1585. [PMID: 31836609 DOI: 10.1158/1078-0432.ccr-19-3053] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE The STAKT study examined short-term exposure (4.5 days) to the oral selective pan-AKT inhibitor capivasertib (AZD5363) to determine if this drug can reach its therapeutic target in sufficient concentration to significantly modulate key biomarkers of the AKT pathway and tumor proliferation. PATIENTS AND METHODS STAKT was a two-stage, double-blind, randomized, placebo-controlled, "window-of-opportunity" study in patients with newly diagnosed ER+ invasive breast cancer. Stage 1 assessed capivasertib 480 mg b.i.d. (recommended monotherapy dose) and placebo, and stage 2 assessed capivasertib 360 and 240 mg b.i.d. Primary endpoints were changes from baseline in AKT pathway markers pPRAS40, pGSK3β, and proliferation protein Ki67. Pharmacologic and pharmacodynamic properties were analyzed from blood sampling, and tolerability by adverse-event monitoring. RESULTS After 4.5 days' exposure, capivasertib 480 mg b.i.d. (n = 17) produced significant decreases from baseline versus placebo (n = 11) in pGSK3β (H-score absolute change: -55.3, P = 0.006) and pPRAS40 (-83.8, P < 0.0001), and a decrease in Ki67 (absolute change in percentage positive nuclei: -9.6%, P = 0.031). Significant changes also occurred in secondary signaling biomarker pS6 (-42.3, P = 0.004), while pAKT (and nuclear FOXO3a) also increased in accordance with capivasertib's mechanism (pAKT: 81.3, P = 0.005). At doses of 360 mg b.i.d. (n = 5) and 240 mg b.i.d. (n = 6), changes in primary and secondary biomarkers were also observed, albeit of smaller magnitude. Biomarker modulation was dose and concentration dependent, and no new safety signals were evident. CONCLUSIONS Capivasertib 480 mg b.i.d. rapidly modulates key biomarkers of the AKT pathway and decreases proliferation marker Ki67, suggesting future potential as an effective therapy in AKT-dependent breast cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Anthony Skene
- Royal Bournemouth and Christchurch Hospitals NHS Foundation Trust, Bournemouth, UK
| | - Daniel Rea
- University of Birmingham, Birmingham, UK
| | | | - Ali Jahan
- King's Mill Hospital, Nottingham, UK
| | | | | | | | | | | | | | | | | | - Martin Pass
- IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Paul Rugman
- IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | - Rahul Deb
- Department of Histopathology, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, UK
| | | | | | | |
Collapse
|
6
|
Gougis P, Moreau Bachelard C, Kamal M, Gan HK, Borcoman E, Torossian N, Bièche I, Le Tourneau C. Clinical Development of Molecular Targeted Therapy in Head and Neck Squamous Cell Carcinoma. JNCI Cancer Spectr 2019; 3:pkz055. [PMID: 32337482 PMCID: PMC7049986 DOI: 10.1093/jncics/pkz055] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/12/2019] [Accepted: 07/26/2019] [Indexed: 12/26/2022] Open
Abstract
A better understanding of cancer biology has led to the development of molecular targeted therapy, which has dramatically improved the outcome of some cancer patients, especially when a biomarker of efficacy has been used for patients' selection. In head and neck oncology, cetuximab that targets epidermal growth factor receptor is the only targeted therapy that demonstrated a survival benefit, both in the recurrent and in the locally advanced settings, yet without prior patients' selection. We herein review the clinical development of targeted therapy in head and neck squamous cell carcinoma in light of the molecular landscape and give insights in on how innovative clinical trial designs may speed up biomarker discovery and deployment of new molecular targeted therapies. Given the recent approval of immune checkpoint inhibitors targeting programmed cell death-1 in head and neck squamous cell carcinoma, it remains to be determined how targeted therapy will be incorporated into a global drug development strategy that will inevitably incorporate immunotherapy.
Collapse
Affiliation(s)
- Paul Gougis
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris & Saint-Cloud, France
- AP-HP, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1421, CLIP Galilée, Paris, France
| | - Camille Moreau Bachelard
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris & Saint-Cloud, France
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris & Saint-Cloud, France
| | - Hui K Gan
- Cancer Clinical Trial Centre, Austin Hospital, Heidelberg, Melbourne, Australia
| | - Edith Borcoman
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris & Saint-Cloud, France
| | - Nouritza Torossian
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris & Saint-Cloud, France
| | - Ivan Bièche
- Pharmacogenomics Unit, Institut Curie, Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Paris & Saint-Cloud, France
- INSERM U900 Research Unit, Saint-Cloud, France
- Paris-Saclay University, Paris, France
| |
Collapse
|
7
|
Chen J, Easwaralingam N, Warrier S, Ong A, Carson EK, Mak C, Snook K, Middleton K, Parker A, Palmieri C, Spillane A, Mann GB, Lim E, Segara D. Window of opportunity treatment in breast cancer. ANZ J Surg 2019; 90:34-40. [PMID: 31770829 DOI: 10.1111/ans.15487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 11/27/2022]
Abstract
Window of opportunity therapies, which involve short-term administration of systemic therapy between cancer diagnosis and surgery, have raised significant interest in recent years as a mean of assessing the sensitivity of a patient's cancer to therapy prior to surgery. There is now compelling evidence that in patients with early stage hormone-receptor positive breast cancer, a 2-week preoperative treatment with standard hormone therapies in a preoperative window period provides important prognostic information, which in turn helps to aid decision-making regarding treatment options. Changes in short-term biomarker endpoints such as cell proliferation measured by Ki-67 can act as surrogate markers of long-term outcomes. Paired tissues obtained pre- and post-investigational treatment, without having to subject the patient to additional biopsies, can then be used to conduct translational research to investigate predictive biomarkers and pharmacodynamics. In this review, we will examine the utility and challenges of window of opportunities therapies in breast cancer in the current literature, and the current Australian and international trial landscape in this clinical space.
Collapse
Affiliation(s)
- Julia Chen
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Neshanth Easwaralingam
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Sanjay Warrier
- Chris O'Brien Lifehouse and The University of Sydney, Sydney, New South Wales, Australia.,Royal Prince Alfred Hospital Institute of Academic Surgery, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew Ong
- Campbelltown Hospital and The University of Western Sydney, Sydney, New South Wales, Australia
| | - Emma-Kate Carson
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia.,Campbelltown Hospital and The University of Western Sydney, Sydney, New South Wales, Australia
| | - Cindy Mak
- Chris O'Brien Lifehouse and The University of Sydney, Sydney, New South Wales, Australia
| | - Kylie Snook
- Breast and Surgical Oncology at the Poche Centre and Northern Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Kate Middleton
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Andrew Parker
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Carlo Palmieri
- University of Liverpool, Clatterbridge Cancer Centre, NHS Foundation Trust, and Royal Liverpool University Hospital, Liverpool, UK
| | - Andrew Spillane
- Breast and Surgical Oncology at the Poche Centre and Northern Clinical School, The University of Sydney, Sydney, New South Wales, Australia.,Mater Hospital, Sydney, New South Wales, Australia.,Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - G Bruce Mann
- The Royal Melbourne Hospital and The Royal Women's Hospital, Melbourne, Victoria, Australia.,Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Elgene Lim
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| | - Davendra Segara
- Garvan Institute of Medical Research and St Vincent's Hospital, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Aspeslagh S, Morel D, Soria JC, Postel-Vinay S. Epigenetic modifiers as new immunomodulatory therapies in solid tumours. Ann Oncol 2019; 29:812-824. [PMID: 29432557 DOI: 10.1093/annonc/mdy050] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Immune therapies have revolutionized cancer treatment over the last few years by allowing improvements in overall survival. However, the majority of patients is still primary or secondary resistant to such therapies, and enhancing sensitivity to immune therapies is therefore crucial to improve patient outcome. Several recent lines of evidence suggest that epigenetic modifiers have intrinsic immunomodulatory properties, which could be of therapeutic interest. Material and methods We reviewed preclinical evidence and clinical studies which describe or exploit immunomodulatory properties of epigenetic agents. Experimental approaches, clinical applicability and corresponding ongoing clinical trials are described. Results Several epigenetic modifiers, such as histone deacetylase inhibitors, DNA methyl transferase inhibitors, bromodomain inhibitors, lysine-specific histone demethylase 1 inhibitors and enhancer of zeste homolog 2 inhibitors, display intrinsic immunomodulatory properties. The latter can be achieved through the action of these drugs either on cancer cells (e.g. presentation and generation of neoantigens, induction of immunogenic cell death, modulation of cytokine secretion), on immune cells (e.g. linage, differentiation, activation status and antitumor capability), or on components of the microenvironment (e.g. regulatory T cells and macrophages). Several promising combinations, notably with immune checkpoint blockers or adoptive T-cell therapy, can be envisioned. Dedicated clinically relevant approaches for patient selection and trial design will be required to optimally develop such combinations. Conclusion In an era where immune therapies are becoming a treatment backbone in many tumour types, epigenetic modifiers could play a crucial role in modulating tumours' immunogenicity and sensitivity to immune agents. Optimal trial design, including window of opportunity trials, will be key in the success of this approach, and clinical evaluation is ongoing.
Collapse
Affiliation(s)
- S Aspeslagh
- Department of Medical Oncology, Institut Jules Bordet - ULB, Brussels, Belgium
| | - D Morel
- INSERM, UMR981, Villejuif, France
| | - J-C Soria
- INSERM, UMR981, Villejuif, France; Drug Development Department (DITEP, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - S Postel-Vinay
- INSERM, UMR981, Villejuif, France; Drug Development Department (DITEP, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France.
| |
Collapse
|
9
|
Vendrell JA, Solassol J, Győrffy B, Vilquin P, Jarlier M, Donini CF, Gamba L, Maudelonde T, Rouanet P, Cohen PA. Evaluating ZNF217 mRNA Expression Levels as a Predictor of Response to Endocrine Therapy in ER+ Breast Cancer. Front Pharmacol 2019; 9:1581. [PMID: 30740056 PMCID: PMC6355665 DOI: 10.3389/fphar.2018.01581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/31/2018] [Indexed: 11/13/2022] Open
Abstract
ZNF217 is a candidate oncogene with a wide variety of deleterious functions in breast cancer. Here, we aimed at investigating in a pilot prospective study the association between ZNF217 mRNA expression levels and the clinical response to neoadjuvant endocrine therapy (ET) in postmenopausal ER-positive (ER+) breast cancer patients. Core surgical biopsy samples before treatment initiation and post-treatment were obtained from 68 patients, and Ki-67 values measured by immunohistochemistry (IHC) were used to identify responders (n = 59) and non-responders (n = 9) after 4 months of ET. We report for the first time that high ZNF217 mRNA expression level measured by RT-qPCR in the initial tumor samples (pre-treatment) is associated with poor response to neoadjuvant ET. Indeed, the clinical positive response rate in patients with low ZNF217 expression levels was significantly higher than that in those with high ZNF217 expression levels (P = 0.027). Additionally, a retrospective analysis evaluating ZNF217 expression levels in primary breast tumor of ER+/HER2-/LN0 breast cancer patients treated with adjuvant ET enabled the identification of poorer responders prone to earlier relapse (P = 0.013), while ZNF217 did not retain any prognostic value in the ER+/HER2-/LN0 breast cancer patients who did not receive any treatment. Altogether, these data suggest that ZNF217 expression might be predictive of clinical response to ET.
Collapse
Affiliation(s)
- Julie A Vendrell
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jérôme Solassol
- Département de Pathologie et Oncobiologie, Laboratoire de Biologie des Tumeurs Solides, CHU Montpellier, University of Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Montpellier, France
| | - Balázs Győrffy
- 2nd Department of Paediatrics, Semmelweis University, Budapest, Hungary.,MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Budapest, Hungary
| | - Paul Vilquin
- Département de Pathologie et Oncobiologie, Laboratoire de Biologie des Tumeurs Solides, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Marta Jarlier
- Biometrics Unit, Institut du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Caterina F Donini
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Laurent Gamba
- Département de Pathologie et Oncobiologie, Laboratoire de Biologie des Tumeurs Solides, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Thierry Maudelonde
- Département de Pathologie et Oncobiologie, Laboratoire de Biologie des Tumeurs Solides, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Philippe Rouanet
- Département de Chirurgie Oncologique, Institut du Cancer de Montpellier, University of Montpellier, Montpellier, France
| | - Pascale A Cohen
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| |
Collapse
|
10
|
Akcakanat A, Meric-Bernstam F. MK-2206 window of opportunity study in breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:S57. [PMID: 30613632 DOI: 10.21037/atm.2018.10.32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Argun Akcakanat
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Tanić M, Krivokuća A, Čavić M, Mladenović J, Plesinac Karapandžić V, Beck S, Radulović S, Susnjar S, Janković R. Molecular signature of response to preoperative radiotherapy in locally advanced breast cancer. Radiat Oncol 2018; 13:193. [PMID: 30285791 PMCID: PMC6167820 DOI: 10.1186/s13014-018-1129-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 09/12/2018] [Indexed: 01/07/2023] Open
Abstract
Background Radiation therapy is an indispensable part of various treatment modalities for breast cancer. Specifically, for non-inflammatory locally advanced breast cancer (LABC) patients, preoperative radiotherapy (pRT) is currently indicated as a second line therapy in the event of lack of response to neoadjuvant chemotherapy. Still approximately one third of patients fails to respond favourably to pRT. The aim of this study was to explore molecular mechanisms underlying differential response to radiotherapy (RT) to identify predictive biomarkers and potential targets for increasing radiosensitivity. Methods The study was based on a cohort of 134 LABC patients, treated at the Institute of Oncology and Radiology of Serbia (IORS) with pRT, without previous or concomitant systemic therapy. Baseline transcriptional profiles were established using Agilent 60 K microarray platform in a subset of 23 formalin-fixed paraffin-embedded (FFPE) LABC tumour samples of which 11 radiotherapy naïve and 3 post-radiotherapy samples passed quality control and were used for downstream analysis. Biological networks and signalling pathways underlying differential response to RT were identified using Ingenuity Pathways Analysis software. Predictive value of candidate genes in the preoperative setting was further validated by qRT-PCR in an independent subset of 60 LABC samples of which 42 had sufficient quality for data analysis, and in postoperative setting using microarray data from 344 node-negative breast cancer patients (Erasmus cohort, GSE2034 and GSE5327) treated either with surgery only (20%) or surgery with RT (80%). Results We identified 192 significantly differentially expressed genes (FDR < 0.10) between pRT-responsive and non-responsive tumours, related to regulation of cellular development, growth and proliferation, cell cycle control of chromosomal replication, glucose metabolism and NAD biosynthesis II route. APOA1, MAP3K4, and MMP14 genes were differentially expressed (FDR < 0.20) between pRT responders and non-responders in preoperative setting, while MAP3K4 was further validated as RT-specific predictive biomarker of distant metastasis free survival (HR = 2.54, [95%CI:1.42–4.55], p = 0.002) in the postoperative setting. Conclusions This study pinpoints MAP3K4 as a putative biomarker of response to RT in both preoperative and postoperative settings and a potential target for radiosensitising combination therapy, warranting further pre-clinical studies and prospective clinical validation. Electronic supplementary material The online version of this article (10.1186/s13014-018-1129-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miljana Tanić
- Laboratory for Molecular Genetics, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia. .,Medical Genomics, UCL Cancer Institute, University College London, London, UK.
| | - Ana Krivokuća
- Laboratory for Molecular Genetics, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Milena Čavić
- Laboratory for Molecular Genetics, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jasmina Mladenović
- Radiology and Radiotherapy Department, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | | | - Stephan Beck
- Medical Genomics, UCL Cancer Institute, University College London, London, UK
| | - Siniša Radulović
- Laboratory for Molecular Genetics, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Snezana Susnjar
- Medical Oncology Department, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Radmila Janković
- Laboratory for Molecular Genetics, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| |
Collapse
|
12
|
Machiels JP, Bossi P, Menis J, Lia M, Fortpied C, Liu Y, Lhommel R, Lemort M, Schmitz S, Canevari S, De Cecco L, Guzzo M, Bianchi R, Quattrone P, Crippa F, Duprez T, Lalami Y, Quiriny M, de Saint Aubain N, Clement P, Coropciuc R, Hauben E, Licitra L. Activity and safety of afatinib in a window preoperative EORTC study in patients with squamous cell carcinoma of the head and neck (SCCHN). Ann Oncol 2018; 29:985-991. [DOI: 10.1093/annonc/mdy013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
13
|
Guerrero-Zotano AL, Arteaga CL. Neoadjuvant Trials in ER + Breast Cancer: A Tool for Acceleration of Drug Development and Discovery. Cancer Discov 2017; 7:561-574. [PMID: 28495849 PMCID: PMC5497752 DOI: 10.1158/2159-8290.cd-17-0228] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/03/2017] [Accepted: 04/17/2017] [Indexed: 12/17/2022]
Abstract
Neoadjuvant therapy trials offer an excellent strategy for drug development and discovery in breast cancer, particularly in triple-negative and HER2-overexpressing subtypes, where pathologic complete response is a good surrogate of long-term patient benefit. For estrogen receptor-positive (ER+) breast cancers, however, use of this strategy has been challenging because of the lack of validated surrogates of long-term efficacy and the overall good prognosis of the majority of patients with this cancer subtype. We review below the clinical benefits of neoadjuvant endocrine therapy for ER+/HER2-negative breast cancer, its use and limitations for drug development, prioritization of adjuvant and metastatic trials, and biomarker discovery.Significance: Neoadjuvant endocrine therapy is an excellent platform for the development of investigational drugs, triaging of novel combinations, biomarker validation, and discovery of mechanisms of drug resistance. This review summarizes the clinical and investigational benefits of this approach, with a focus on how to best integrate predictive biomarkers into novel clinical trial designs. Cancer Discov; 7(6); 561-74. ©2017 AACR.
Collapse
Affiliation(s)
- Angel L Guerrero-Zotano
- Departments of Medicine and Cancer Biology; Breast Cancer Program, Vanderbilt-Ingram Cancer Center; Vanderbilt University Medical Center, Nashville, Tennessee
| | - Carlos L Arteaga
- Departments of Medicine and Cancer Biology; Breast Cancer Program, Vanderbilt-Ingram Cancer Center; Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
14
|
Selli C, Dixon JM, Sims AH. Accurate prediction of response to endocrine therapy in breast cancer patients: current and future biomarkers. Breast Cancer Res 2016; 18:118. [PMID: 27903276 PMCID: PMC5131493 DOI: 10.1186/s13058-016-0779-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Approximately 70% of patients have breast cancers that are oestrogen receptor alpha positive (ER+) and are therefore candidates for endocrine treatment. Many of these patients relapse in the years during or following completion of adjuvant endocrine therapy. Thus, many ER+ cancers have primary resistance or develop resistance to endocrine therapy during treatment. Recent improvements in our understanding of how tumours evolve during treatment with endocrine agents have identified both changes in gene expression and mutational profiles, in the primary cancer as well as in circulating tumour cells. Analysing these changes has the potential to improve the prediction of which specific patients will respond to endocrine treatment. Serially profiled biopsies during treatment in the neoadjuvant setting offer promise for accurate and early prediction of response to both current and novel drugs and allow investigation of mechanisms of resistance. In addition, recent advances in monitoring tumour evolution through non-invasive (liquid) sampling of circulating tumour cells and cell-free tumour DNA may provide a method to detect resistant clones and allow implementation of personalized treatments for metastatic breast cancer patients. This review summarises current and future biomarkers and signatures for predicting response to endocrine treatment, and discusses the potential for using approved drugs and novel agents to improve outcomes. Increased prediction accuracy is likely to require sequential sampling, utilising preoperative or neoadjuvant treatment and/or liquid biopsies and an improved understanding of both the dynamics and heterogeneity of breast cancer.
Collapse
Affiliation(s)
- Cigdem Selli
- Applied Bioinformatics of Cancer, Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK.,Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, 35050, Turkey
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, EH4 2XR, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK.
| |
Collapse
|
15
|
Window of opportunity studies: Do they fulfil our expectations? Cancer Treat Rev 2016; 43:50-7. [DOI: 10.1016/j.ctrv.2015.12.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/21/2015] [Accepted: 12/22/2015] [Indexed: 11/21/2022]
|