1
|
Karajannis MA, Onar-Thomas A, Lin T, Baxter PA, Boué DR, Cole BL, Fuller C, Haque S, Jabado N, Lucas JT, MacDonald SM, Matsushima C, Patel N, Pierson CR, Souweidane MM, Thomas DL, Walsh MF, Zaky W, Leary SES, Gajjar A, Fouladi M, Cohen KJ. Phase 2 trial of veliparib, local irradiation, and temozolomide in patients with newly diagnosed high-grade glioma: a Children's Oncology Group study. Neuro Oncol 2025; 27:1092-1101. [PMID: 39560182 PMCID: PMC12083075 DOI: 10.1093/neuonc/noae247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND The outcome for pediatric patients with high-grade glioma (HGG) remains poor. Veliparib, a potent oral poly(adenosine diphosphate-ribose) polymerase (PARP) 1/2 inhibitor, enhances the activity of radiotherapy and DNA-damaging chemotherapy. METHODS We conducted a single-arm, non-randomized phase 2 clinical trial to determine whether treatment with veliparib and radiotherapy, followed by veliparib and temozolomide, improves progression-free survival in pediatric patients with newly diagnosed HGG without H3 K27M or BRAF mutations, compared to patient-level data from historical cohorts with closely matching clinical and molecular features. Following surgical resection, newly diagnosed children with non-metastatic HGG were screened by rapid central pathology review and molecular testing. Eligible patients were enrolled on Stratum 1 (IDH wild-type) or Stratum 2 (IDH mutant). RESULTS Both strata were closed to accrual for futility after planned interim analyses. Among the 23 eligible patients who enrolled on Stratum 1 and received protocol therapy, the 1-year event-free survival (EFS) was 23% (standard error, SE = 9%) and the 1-year overall survival (OS) was 64% (SE = 10%). Among the 14 eligible patients who enrolled on Stratum 2 and received protocol therapy, the 1-year EFS was 57% (SE = 13%) and 1-year OS was 93% (SE = 0.7%). CONCLUSIONS Rapid central pathology review and molecular testing for eligibility were feasible. The protocol therapy including radiation, veliparib, and temozolomide was well tolerated but failed to improve outcomes compared to clinically and molecularly matched historical control cohorts treated with higher doses of alkylator chemotherapy. CLINICALTRIALS.GOV IDENTIFIER NCT03581292 (first posted: July 10, 2018).
Collapse
Affiliation(s)
- Matthias A Karajannis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Tong Lin
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Patricia A Baxter
- Department of Pediatrics, Texas Children’s Hospital/Baylor College of Medicine, Houston, Texas
| | - Daniel R Boué
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Bonnie L Cole
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Christine Fuller
- Department of Pathology, Upstate Medical University, Syracuse, New York, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nada Jabado
- Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| | - John T Lucas
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Shannon M MacDonald
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Celeste Matsushima
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Namrata Patel
- Department of Pharmacy, Stanford Medicine Children’s Health, Palo Alto, California, USA
| | - Christopher R Pierson
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
- Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | - Diana L Thomas
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Michael F Walsh
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Wafik Zaky
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarah E S Leary
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Amar Gajjar
- Department of Pediatric Medicine, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Maryam Fouladi
- Department of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Kenneth J Cohen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
2
|
Rodgers LT, Villano JL, Hartz AMS, Bauer B. Glioblastoma Standard of Care: Effects on Tumor Evolution and Reverse Translation in Preclinical Models. Cancers (Basel) 2024; 16:2638. [PMID: 39123366 PMCID: PMC11311277 DOI: 10.3390/cancers16152638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Glioblastoma (GBM) presents a significant public health challenge as the deadliest and most common malignant brain tumor in adults. Despite standard-of-care treatment, which includes surgery, radiation, and chemotherapy, mortality rates are high, underscoring the critical need for advancing GBM therapy. Over the past two decades, numerous clinical trials have been performed, yet only a small fraction demonstrated a benefit, raising concerns about the predictability of current preclinical models. Traditionally, preclinical studies utilize treatment-naïve tumors, failing to model the clinical scenario where patients undergo standard-of-care treatment prior to recurrence. Recurrent GBM generally exhibits distinct molecular alterations influenced by treatment selection pressures. In this review, we discuss the impact of treatment-surgery, radiation, and chemotherapy-on GBM. We also provide a summary of treatments used in preclinical models, advocating for their integration to enhance the translation of novel strategies to improve therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Louis T. Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - John L. Villano
- Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Anika M. S. Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
3
|
Liu Z, Yang L, Xie Z, Yu H, Gu T, Shi D, Cai N, Zhuo S. Multi-cohort comprehensive analysis unveiling the clinical value and therapeutic effect of GNAL in glioma. Oncol Res 2024; 32:965-981. [PMID: 38686055 PMCID: PMC11055992 DOI: 10.32604/or.2024.045769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/21/2023] [Indexed: 05/02/2024] Open
Abstract
Clinical data indicates that glioma patients have poor treatment outcomes and clinical prognosis. The role of olfactory signaling pathway-related genes (OSPRGs) in glioma has not been fully elucidated. In this study, we aimed to investigate the role and relationship between OSPRGs and glioma. Univariate and multivariate Cox regression analyses were performed to assess the relationship between OSPRGs and the overall survival of glioma based on public cohorts, and the target gene (G Protein Subunit Alpha L, GNAL) was screened. The association of GNAL expression with clinicopathological characteristics, gene mutation landscape, tumor immune microenvironment (TIME), deoxyribonucleic acid (DNA) methylation, and naris-occlusion controlled genes (NOCGs) was performed. Immunohistochemistry was used to evaluate GNAL level in glioma. Further analysis was conducted to evaluate the drug sensitivity, immunotherapy response, and functional enrichment of GNAL. GNAL was an independent prognostic factor, and patients with low GNAL expression have a poor prognosis. Expression of GNAL was closely associated with clinicopathological characteristics, DNA methylation, and several immune-related pathways. Immune infiltration analysis indicated that GNAL levels were negatively correlated with immune scores. GNAL low-expression group showed efficacy with anti-PD-1 therapy. Ten compounds with significantly different half-maximal inhibitory concentration (IC50) values between the GNAL high and low-expression groups were identified. Furthermore, its expression was associated with several immune cells, immune-related genes, and NOCGs. The expression of GNAL is closely associated with clinicopathological characteristics, TIME, and the response to therapeutic interventions, highlighting its potential as a prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Liangwang Yang
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Zhengxing Xie
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Hui Yu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Tianyi Gu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Daoming Shi
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Ning Cai
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Shenghua Zhuo
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| |
Collapse
|
4
|
Perspective on the Use of DNA Repair Inhibitors as a Tool for Imaging and Radionuclide Therapy of Glioblastoma. Cancers (Basel) 2022; 14:cancers14071821. [PMID: 35406593 PMCID: PMC8997380 DOI: 10.3390/cancers14071821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/03/2023] Open
Abstract
Simple Summary The current routine treatment for glioblastoma (GB), the most lethal high-grade brain tumor in adults, aims to induce DNA damage in the tumor. However, the tumor cells might be able to repair that damage, which leads to therapy resistance. Fortunately, DNA repair defects are common in GB cells, and their survival is often based on a sole backup repair pathway. Hence, targeted drugs inhibiting essential proteins of the DNA damage response have gained momentum and are being introduced in the clinic. This review gives a perspective on the use of radiopharmaceuticals targeting DDR kinases for imaging in order to determine the DNA repair phenotype of GB, as well as for effective radionuclide therapy. Finally, four new promising radiopharmaceuticals are suggested with the potential to lead to a more personalized GB therapy. Abstract Despite numerous innovative treatment strategies, the treatment of glioblastoma (GB) remains challenging. With the current state-of-the-art therapy, most GB patients succumb after about a year. In the evolution of personalized medicine, targeted radionuclide therapy (TRT) is gaining momentum, for example, to stratify patients based on specific biomarkers. One of these biomarkers is deficiencies in DNA damage repair (DDR), which give rise to genomic instability and cancer initiation. However, these deficiencies also provide targets to specifically kill cancer cells following the synthetic lethality principle. This led to the increased interest in targeted drugs that inhibit essential DDR kinases (DDRi), of which multiple are undergoing clinical validation. In this review, the current status of DDRi for the treatment of GB is given for selected targets: ATM/ATR, CHK1/2, DNA-PK, and PARP. Furthermore, this review provides a perspective on the use of radiopharmaceuticals targeting these DDR kinases to (1) evaluate the DNA repair phenotype of GB before treatment decisions are made and (2) induce DNA damage via TRT. Finally, by applying in-house selection criteria and analyzing the structural characteristics of the DDRi, four drugs with the potential to become new therapeutic GB radiopharmaceuticals are suggested.
Collapse
|
5
|
Parsels LA, Zhang Q, Karnak D, Parsels JD, Lam K, Willers H, Green MD, Rehemtulla A, Lawrence TS, Morgan MA. Translation of DNA Damage Response Inhibitors as Chemoradiation Sensitizers From the Laboratory to the Clinic. Int J Radiat Oncol Biol Phys 2021; 111:e38-e53. [PMID: 34348175 PMCID: PMC8602768 DOI: 10.1016/j.ijrobp.2021.07.1708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022]
Abstract
Combination therapies with agents targeting the DNA damage response (DDR) offer an opportunity to selectively enhance the therapeutic index of chemoradiation or eliminate use of chemotherapy altogether. The successful translation of DDR inhibitors to clinical use requires investigating both their direct actions as (chemo)radiosensitizers and their potential to stimulate tumor immunogenicity. Beginning with high-throughput screening using both viability and DNA damage-reporter assays, followed by validation in gold-standard radiation colony-forming assays and in vitro assessment of mechanistic effects on the DDR, we describe proven strategies and methods leading to the clinical development of DDR inhibitors both with radiation alone and in combination with chemoradiation. Beyond these in vitro studies, we discuss the impact of key features of human xenograft and syngeneic mouse models on the relevance of in vivo tumor efficacy studies, particularly with regard to the immunogenic effects of combined therapy with radiation and DDR inhibitors. Finally, we describe recent technological advances in radiation delivery (using the small animal radiation research platform) that allow for conformal, clinically relevant radiation therapy in mouse models. This overall approach is critical to the successful clinical development and ultimate Food and Drug Administration approval of DDR inhibitors as (chemo)radiation sensitizers.
Collapse
Affiliation(s)
- Leslie A Parsels
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Qiang Zhang
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - David Karnak
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Joshua D Parsels
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Kwok Lam
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael D Green
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, Ann Arbor, Michigan.
| |
Collapse
|
6
|
Ulasov IV, Borovjagin A, Laevskaya A, Kamynina M, Timashev P, Cerchia L, Rozhkova EA. The IL13α 2R paves the way for anti-glioma nanotherapy. Genes Dis 2021; 10:89-100. [PMID: 37013057 PMCID: PMC10066331 DOI: 10.1016/j.gendis.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive (grade IV) gliomas characterized by a high rate of recurrence, resistance to therapy and a grim survival prognosis. The long-awaited improvement in GBM patients' survival rates essentially depends on advances in the development of new therapeutic approaches. Recent preclinical studies show that nanoscale materials could greatly contribute to the improvement of diagnosis and management of brain cancers. In the current review, we will discuss how specific features of glioma pathobiology can be employed for designing efficient targeting approaches. Moreover, we will summarize the main evidence for the potential of the IL-13R alpha 2 receptor (IL13α2R) targeting in GBM early diagnosis and experimental therapy.
Collapse
Affiliation(s)
- Ilya V. Ulasov
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Corresponding author.
| | - Anton Borovjagin
- Department of BioMedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anastasia Laevskaya
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Margarita Kamynina
- Group of Experimental Biotherapy and Diagnostic, Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 8-2 Trubetskaya St., Moscow 119991, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, 4 Kosygin St, Moscow 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, Leninskiye Gory 1-3, Moscow 119991, Russia
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), Naples 80131, Italy
| | - Elena A. Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, Argonne, IL 60439, USA
| |
Collapse
|
7
|
Inhibition of DNA Repair in Combination with Temozolomide or Dianhydrogalactiol Overcomes Temozolomide-Resistant Glioma Cells. Cancers (Basel) 2021; 13:cancers13112570. [PMID: 34073837 PMCID: PMC8197190 DOI: 10.3390/cancers13112570] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Glioblastoma is the most prevalent and lethal brain tumor. Temozolomide is usually used for the treatment of glioblastoma. The poor prognosis of the tumor is due to drug resistance and tumor heterogeneity. The mechanism of the resistance to temozolomide is various within the same tumor. The aim of the study was to clarify the mechanism of temozolomide resistance and find methods to overcome temozolomide resistance in glioma. Inhibition of DNA repair (homologous recombination or base excision repair) resensitized resistant cells harboring different resistance mechanism to temozolomide. Additionally, a bifunctional DNA-targeting agent, dianhydrogalactiol, showed anti-tumor effect independent of MGMT and mismatch repair status. Further, inhibition of checkpoint or homologous recombination enhanced dianhydrogalactiol-induced cytotoxicity in temozolomide-resistant glioma cells. Although resistance to temozolomide is clinically important issue, selecting suitable treatments for resistance mechanism can improve the prognosis of glioma. Abstract Resistance to temozolomide and intratumoral heterogeneity contribute to the poor prognosis of glioma. The mechanisms of temozolomide resistance can vary within a heterogeneous tumor. Temozolomide adds a methyl group to DNA. The primary cytotoxic lesion, O6-methylguanine, mispairs with thymine, leading to a futile DNA mismatch repair cycle, formation of double-strand breaks, and eventual cell death when O6-methylguanine DNA methyltransferase (MGMT) is absent. N7-methylguanine and N3-methyladenine are repaired by base excision repair (BER). The study aim was to elucidate temozolomide resistance mechanisms and identify methods to overcome temozolomide resistance in glioma. Several temozolomide-resistant clones were analyzed. Increased homologous recombination and mismatch repair system deficiencies contributed to temozolomide resistance. Inhibition of homologous recombination resensitized resistant cells with high homologous recombination efficiency. For the mismatch repair-deficient cells, inhibition of BER by PARP inhibitor potentiated temozolomide-induced cytotoxicity. Dianhydrogalactiol is a bifunctional DNA-targeting agent that forms N7-alkylguanine and inter-strand DNA crosslinks. Dianhydrogalactiol reduced the proliferation of cells independent of MGMT and mismatch repair, inducing DNA double-strand breaks and apoptosis in temozolomide-resistant cells. Further, inhibition of chk1 or homologous recombination enhanced dianhydrogalactiol-induced cytotoxicity in the cells. Selecting treatments most appropriate to the types of resistance mechanisms can potentially improve the prognosis of glioma.
Collapse
|
8
|
Baxter PA, Su JM, Onar-Thomas A, Billups CA, Li XN, Poussaint TY, Smith ER, Thompson P, Adesina A, Ansell P, Giranda V, Paulino A, Kilburn L, Quaddoumi I, Broniscer A, Blaney SM, Dunkel IJ, Fouladi M. A phase I/II study of veliparib (ABT-888) with radiation and temozolomide in newly diagnosed diffuse pontine glioma: a Pediatric Brain Tumor Consortium study. Neuro Oncol 2021; 22:875-885. [PMID: 32009149 DOI: 10.1093/neuonc/noaa016] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A Pediatric Brain Tumor Consortium (PBTC) phase I/II trial of veliparib and radiation followed by veliparib and temozolomide (TMZ) was conducted in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). The objectives were to: (i) estimate the recommended phase II dose (RP2D) of veliparib with concurrent radiation; (ii) evaluate the pharmacokinetic parameters of veliparib during radiation; (iii) evaluate feasibility of intrapatient TMZ dose escalation; (iv) describe toxicities of protocol therapy; and (v) estimate the overall survival distribution compared with historical series. METHODS Veliparib was given Monday through Friday b.i.d. during radiation followed by a 4-week rest. Patients then received veliparib at 25 mg/m2 b.i.d. and TMZ 135 mg/m2 daily for 5 days every 28 days. Intrapatient dose escalation of TMZ was investigated for patients experiencing minimal toxicity. RESULTS Sixty-six patients (65 eligible) were enrolled. The RP2D of veliparib was 65 mg/m2 b.i.d. with radiation. Dose-limiting toxicities during radiation with veliparib therapy included: grade 2 intratumoral hemorrhage (n = 1), grade 3 maculopapular rash (n = 2), and grade 3 nervous system disorder (generalized neurologic deterioration) (n = 1). Intrapatient TMZ dose escalation during maintenance was not tolerated. Following a planned interim analysis, it was concluded that this treatment did not show a survival benefit compared with PBTC historical controls, and accrual was stopped for futility. The 1- and 2-year overall survival rates were 37.2% (SE 7%) and 5.3% (SE 3%), respectively. CONCLUSION Addition of veliparib to radiation followed by TMZ and veliparib was tolerated but did not improve survival for patients with newly diagnosed DIPG. TRIAL REGISTRATION NCT01514201.
Collapse
Affiliation(s)
- Patricia A Baxter
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | - Jack M Su
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | | | | | - Xiao-Nan Li
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | | | | | - Patrick Thompson
- University of North Carolina Children's Hospital, Chapel Hill, North Carolina
| | - Adekunle Adesina
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | | | | | - Arnold Paulino
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Susan M Blaney
- Texas Children's Hospital/Baylor College of Medicine, Houston, Texas
| | - Ira J Dunkel
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
9
|
New Avenues in Radiotherapy of Glioblastoma: from Bench to Bedside. Curr Treat Options Neurol 2020. [DOI: 10.1007/s11940-020-00654-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
10
|
Brossard C, Montigon O, Boux F, Delphin A, Christen T, Barbier EL, Lemasson B. MP3: Medical Software for Processing Multi-Parametric Images Pipelines. Front Neuroinform 2020; 14:594799. [PMID: 33304261 PMCID: PMC7701116 DOI: 10.3389/fninf.2020.594799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/14/2020] [Indexed: 01/28/2023] Open
Abstract
This article presents an open source software able to convert, display, and process medical images. It differentiates itself from the existing software by its ability to design complex processing pipelines and to wisely execute them on a large databases. An MP3 pipeline can contain unlimited homemade or ready-made processes and can be carried out with a parallel execution system. As a viewer, MP3 allows display of up to four images together and to draw Regions Of Interest (ROI). Two applications showing the strengths of the software are presented as examples: a preclinical study involving Magnetic Resonance Imaging (MRI) data and a clinical one involving Computed Tomography (CT) images. MP3 is downloadable at https://github.com/nifm-gin/MP3.
Collapse
Affiliation(s)
- Clément Brossard
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,MoGlimaging Network, HTE Program of the French Cancer Plan, Toulouse, France
| | - Olivier Montigon
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France
| | - Fabien Boux
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,University of Grenoble Alpes, Inria, CNRS, G-INP, Grenoble, France
| | - Aurélien Delphin
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France
| | - Thomas Christen
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France
| | - Emmanuel L Barbier
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France
| | - Benjamin Lemasson
- University of Grenoble Alpes, INSERM, U1216, Grenoble Institut des Neurosciences (GIN), Grenoble, France.,MoGlimaging Network, HTE Program of the French Cancer Plan, Toulouse, France
| |
Collapse
|
11
|
Valtorta S, Salvatore D, Rainone P, Belloli S, Bertoli G, Moresco RM. Molecular and Cellular Complexity of Glioma. Focus on Tumour Microenvironment and the Use of Molecular and Imaging Biomarkers to Overcome Treatment Resistance. Int J Mol Sci 2020; 21:E5631. [PMID: 32781585 PMCID: PMC7460665 DOI: 10.3390/ijms21165631] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023] Open
Abstract
This review highlights the importance and the complexity of tumour biology and microenvironment in the progression and therapy resistance of glioma. Specific gene mutations, the possible functions of several non-coding microRNAs and the intra-tumour and inter-tumour heterogeneity of cell types contribute to limit the efficacy of the actual therapeutic options. In this scenario, identification of molecular biomarkers of response and the use of multimodal in vivo imaging and in particular the Positron Emission Tomography (PET) based molecular approach, can help identifying glioma features and the modifications occurring during therapy at a regional level. Indeed, a better understanding of tumor heterogeneity and the development of diagnostic procedures can favor the identification of a cluster of patients for personalized medicine in order to improve the survival and their quality of life.
Collapse
Affiliation(s)
- Silvia Valtorta
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
| | - Daniela Salvatore
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
| | - Paolo Rainone
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate, Italy
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate, Italy
| | - Rosa Maria Moresco
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate, Italy
| |
Collapse
|
12
|
Philippou Y, Sjoberg H, Lamb AD, Camilleri P, Bryant RJ. Harnessing the potential of multimodal radiotherapy in prostate cancer. Nat Rev Urol 2020; 17:321-338. [PMID: 32358562 DOI: 10.1038/s41585-020-0310-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Radiotherapy in combination with androgen deprivation therapy (ADT) is a standard treatment option for men with localized and locally advanced prostate cancer. However, emerging clinical evidence suggests that radiotherapy can be incorporated into multimodality therapy regimens beyond ADT, in combinations that include chemotherapy, radiosensitizing agents, immunotherapy and surgery for the treatment of men with localized and locally advanced prostate cancer, and those with oligometastatic disease, in whom the low metastatic burden in particular might be treatable with these combinations. This multimodal approach is increasingly recognized as offering considerable clinical benefit, such as increased antitumour effects and improved survival. Thus, radiotherapy is becoming a key component of multimodal therapy for many stages of prostate cancer, particularly oligometastatic disease.
Collapse
Affiliation(s)
- Yiannis Philippou
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Hanna Sjoberg
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK
| | - Philip Camilleri
- Oxford Department of Clinical Oncology, Churchill Hospital Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Headington, Oxford, UK
| | - Richard J Bryant
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Headington, Oxford, UK.
- Nuffield Department of Surgical Sciences, University of Oxford, Headington, Oxford, UK.
| |
Collapse
|
13
|
A New Pathway Promotes Adaptation of Human Glioblastoma Cells to Glucose Starvation. Cells 2020; 9:cells9051249. [PMID: 32443613 PMCID: PMC7290719 DOI: 10.3390/cells9051249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/11/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Adaptation of glioblastoma to caloric restriction induces compensatory changes in tumor metabolism that are incompletely known. Here we show that in human glioblastoma cells maintained in exhausted medium, SHC adaptor protein 3 (SHC3) increases due to down-regulation of SHC3 protein degradation. This effect is reversed by glucose addition and is not present in normal astrocytes. Increased SHC3 levels are associated to increased glucose uptake mediated by changes in membrane trafficking of glucose transporters of the solute carrier 2A superfamily (GLUT/SLC2A). We found that the effects on vesicle trafficking are mediated by SHC3 interactions with adaptor protein complex 1 and 2 (AP), BMP-2-inducible protein kinase and a fraction of poly ADP-ribose polymerase 1 (PARP1) associated to vesicles containing GLUT/SLC2As. In glioblastoma cells, PARP1 inhibitor veliparib mimics glucose starvation in enhancing glucose uptake. Furthermore, cytosol extracted from glioblastoma cells inhibits PARP1 enzymatic activity in vitro while immunodepletion of SHC3 from the cytosol significantly relieves this inhibition. The identification of a new pathway controlling glucose uptake in high grade gliomas represents an opportunity for repositioning existing drugs and designing new ones.
Collapse
|
14
|
Vajapeyam S, Brown D, Billups C, Patay Z, Vezina G, Shiroishi MS, Law M, Baxter P, Onar-Thomas A, Fangusaro JR, Dunkel IJ, Poussaint TY. Advanced ADC Histogram, Perfusion, and Permeability Metrics Show an Association with Survival and Pseudoprogression in Newly Diagnosed Diffuse Intrinsic Pontine Glioma: A Report from the Pediatric Brain Tumor Consortium. AJNR Am J Neuroradiol 2020; 41:718-724. [PMID: 32241771 DOI: 10.3174/ajnr.a6499] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/10/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Diffuse intrinsic pontine glioma is a lethal childhood brain cancer with dismal prognosis and MR imaging is the primary methodology used for diagnosis and monitoring. Our aim was to determine whether advanced diffusion, perfusion, and permeability MR imaging metrics predict survival and pseudoprogression in children with newly diagnosed diffuse intrinsic pontine glioma. MATERIALS AND METHODS A clinical trial using the poly (adenosine diphosphate ribose) polymerase (PARP) inhibitor veliparib concurrently with radiation therapy, followed by maintenance therapy with veliparib + temozolomide, in children with diffuse intrinsic pontine glioma was conducted by the Pediatric Brain Tumor Consortium. Standard MR imaging, DWI, dynamic contrast-enhanced perfusion, and DSC perfusion were performed at baseline and approximately every 2 months throughout treatment. ADC histogram metrics of T2-weighted FLAIR and enhancing tumor volume, dynamic contrast-enhanced permeability metrics for enhancing tumors, and tumor relative CBV from DSC perfusion MR imaging were calculated. Baseline values, post-radiation therapy changes, and longitudinal trends for all metrics were evaluated for associations with survival and pseudoprogression. RESULTS Fifty children were evaluable for survival analyses. Higher baseline relative CBV was associated with shorter progression-free survival (P = .02, Q = 0.089) and overall survival (P = .006, Q = 0.055). Associations of higher baseline mean transfer constant from the blood plasma into the extravascular extracellular space with shorter progression-free survival (P = .03, Q = 0.105) and overall survival (P = .03, Q = 0.102) trended toward significance. An increase in relative CBV with time was associated with shorter progression-free survival (P < .001, Q < 0.001) and overall survival (P = .004, Q = 0.043). Associations of longitudinal mean extravascular extracellular volume fraction with progression-free survival (P = .03, Q = 0.104) and overall survival (P = .03, Q = 0.105) and maximum transfer constant from the blood plasma into the extravascular extracellular space with progression-free survival (P = .03, Q = 0.102) trended toward significance. Greater increases with time were associated with worse outcomes. True radiologic progression showed greater post-radiation therapy decreases in mode_ADC_FLAIR compared with pseudoprogression (means, -268.15 versus -26.11, P = .01.) CONCLUSIONS: ADC histogram, perfusion, and permeability MR imaging metrics in diffuse intrinsic pontine glioma are useful in predicting survival and pseudoprogression.
Collapse
Affiliation(s)
- S Vajapeyam
- From the Radiology (S.V., T.Y.P.), Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - D Brown
- DF/HCC Tumor Imaging Metrics Core (D.B.), Massachusetts General Hospital, Boston, Massachusetts
| | | | - Z Patay
- Diagnostic Imaging (Z.P.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - G Vezina
- Radiology (G.V.), Children's National Medical Center, Washington, DC
| | - M S Shiroishi
- Radiology (M.S.S.), Keck Medical Center of USC, Los Angeles, California
| | - M Law
- Neuroscience (M.L.), Monash University, Melbourne, Australia
| | - P Baxter
- Cancer and Hematology Center (P.B.), Texas Children's Hospital, Houston, Texas
| | | | - J R Fangusaro
- Aflac Cancer and Blood Disorders Center (J.R.F.), Children's Healthcare of Atlanta, Atlanta, Georgia
| | - I J Dunkel
- Pediatrics (I.J.D.), Memorial Sloan Kettering Cancer Center, New York, New York
| | - T Y Poussaint
- From the Radiology (S.V., T.Y.P.), Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Yang J, Hu S, Shangguan J, Eresen A, Li Y, Ma Q, Yaghmai V, Benson III AB, Zhang Z. Dinaciclib prolongs survival in the LSL-KrasG12D/+ ; LSL-Trp53R172H/+ ; Pdx-1-Cre (KPC) transgenic murine models of pancreatic ductal adenocarcinoma. Am J Transl Res 2020; 12:1031-1043. [PMID: 32269732 PMCID: PMC7137051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/01/2020] [Indexed: 06/11/2023]
Abstract
Dinaciclib is a small molecule cyclin-dependent kinase inhibitor with the potential to treat multiple cancers. To better understand its cytotoxic action in pancreatic ductal adenocarcinoma (PDAC), we evaluated dinaciclib therapeutic effects in the transgenic mouse model (LSL-KrasG12D/+ ; LSL-Trp53R172H/+ ; Pdx-1-Cre mice; KPC mice). Tumor growth and microenvironment were dynamically monitored by magnetic resonance imaging (MRI). Dinaciclib therapy significantly delayed tumor progression (P < 0.001) and prolonged survival (P = 0.007) in KPC mice. In vitro assays showed that dinaciclib exerted antiproliferative effects on PDAC cells by increasing surface calreticulin expression and release of ATP. Dinaciclib treatment inhibited proliferation and induced apoptosis in KPC tumor as assessed by Ki67 and cleaved caspase 3, respectively. Particularly, the tumor infiltrating CD8+ T cells were increased after dinaciclib treatment in KPC mice. Additionally, the mean apparent diffusion coefficient values of KPC tumor calculated from diffusion weighted MR images were significantly lower after dinaciclib treatment (P = 0.033). These finding suggest that dinaciclib as a single agent can inhibit tumor growth and improve the overall survival in KPC mice.
Collapse
Affiliation(s)
- Jia Yang
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Su Hu
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
- Department of Radiology, The First Affiliated Hospital of Soochow UniversitySuzhou, Jiangsu, China
| | - Junjie Shangguan
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Aydin Eresen
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Yu Li
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
- Department of General Surgery, The Affiliated Hospital of Qingdao UniversityQingdao, Shandong, China
| | - Quanhong Ma
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Vahid Yaghmai
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern UniversityChicago, IL, USA
| | - Al B Benson III
- Robert H. Lurie Comprehensive Cancer Center, Northwestern UniversityChicago, IL, USA
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of MedicineChicago, IL, USA
| | - Zhuoli Zhang
- Department of Radiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern UniversityChicago, IL, USA
| |
Collapse
|
16
|
Wu W, Klockow JL, Mohanty S, Ku KS, Aghighi M, Melemenidis S, Chen Z, Li K, Morais GR, Zhao N, Schlegel J, Graves EE, Rao J, Loadman PM, Falconer RA, Mukherjee S, Chin FT, Daldrup-Link HE. Theranostic nanoparticles enhance the response of glioblastomas to radiation. Nanotheranostics 2019; 3:299-310. [PMID: 31723547 PMCID: PMC6838141 DOI: 10.7150/ntno.35342] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 09/14/2019] [Indexed: 01/03/2023] Open
Abstract
Despite considerable progress with our understanding of glioblastoma multiforme (GBM) and the precise delivery of radiotherapy, the prognosis for GBM patients is still unfavorable with tumor recurrence due to radioresistance being a major concern. We recently developed a cross-linked iron oxide nanoparticle conjugated to azademethylcolchicine (CLIO-ICT) to target and eradicate a subpopulation of quiescent cells, glioblastoma initiating cells (GICs), which could be a reason for radioresistance and tumor relapse. The purpose of our study was to investigate if CLIO-ICT has an additive therapeutic effect to enhance the response of GBMs to ionizing radiation. Methods: NSG™ mice bearing human GBMs and C57BL/6J mice bearing murine GBMs received CLIO-ICT, radiation, or combination treatment. The mice underwent pre- and post-treatment magnetic resonance imaging (MRI) scans, bioluminescence imaging (BLI), and histological analysis. Tumor nanoparticle enhancement, tumor flux, microvessel density, GIC, and apoptosis markers were compared between different groups using a one-way ANOVA and two-tailed Mann-Whitney test. Additional NSG™ mice underwent survival analyses with Kaplan-Meier curves and a log rank (Mantel-Cox) test. Results: At 2 weeks post-treatment, BLI and MRI scans revealed significant reduction in tumor size for CLIO-ICT plus radiation treated tumors compared to monotherapy or vehicle-treated tumors. Combining CLIO-ICT with radiation therapy significantly decreased microvessel density, decreased GICs, increased caspase-3 expression, and prolonged the survival of GBM-bearing mice. CLIO-ICT delivery to GBM could be monitored with MRI. and was not significantly different before and after radiation. There was no significant caspase-3 expression in normal brain at therapeutic doses of CLIO-ICT administered. Conclusion: Our data shows additive anti-tumor effects of CLIO-ICT nanoparticles in combination with radiotherapy. The combination therapy proposed here could potentially be a clinically translatable strategy for treating GBMs.
Collapse
Affiliation(s)
- Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Jessica L Klockow
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Suchismita Mohanty
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Kimberly S Ku
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Maryam Aghighi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | | | - Zixin Chen
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Kai Li
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Goreti Ribeiro Morais
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Ning Zhao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Jürgen Schlegel
- Department of Neuropathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Edward E Graves
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA.,Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Paul M Loadman
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Robert A Falconer
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Sudip Mukherjee
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Frederick T Chin
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA, USA
| |
Collapse
|
17
|
Przybycinski J, Nalewajska M, Marchelek-Mysliwiec M, Dziedziejko V, Pawlik A. Poly-ADP-ribose polymerases (PARPs) as a therapeutic target in the treatment of selected cancers. Expert Opin Ther Targets 2019; 23:773-785. [PMID: 31394942 DOI: 10.1080/14728222.2019.1654458] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The implementation of poly-ADP-ribose polymerase (PARP) inhibitors for therapy has created potential treatments for a wide spectrum of malignancies involving DNA damage repair gene abnormalities. PARPs are a group of enzymes that are responsible for detecting and repairing DNA damage and therefore play a key role in maintaining cell function and integrity. PARP inhibitors are drugs that target DNA repair deficiencies. Inhibiting PARP activity in cancer cells causes cell death. Areas covered: This review summarizes the role of PARP inhibitors in the treatment of cancer. We performed a systematic literature search in February 2019 in the electronic databases PubMed and EMBASE. Our search terms were the following: PARP, PARP inhibitors, PARPi, Poly ADP ribose polymerase, cancer treatment. We discuss PARP inhibitors currently being investigated in cancer clinical trials, their safety profiles, clinical resistance, combined therapeutic approaches and future challenges. Expert Opinion: The future could bring novel PARP inhibitors with greater DNA trapping potential, better safety profiles and improved combined therapies involving hormonal, chemo-, radio- or immunotherapies. Progress may afford wider indications for PARP inhibitors in the treatment of cancer and the utilization for cancer prevention in high-risk mutation carriers. Research efforts should focus on identifying novel drugs that target DNA repair deficiencies.
Collapse
Affiliation(s)
- Jarosław Przybycinski
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University , Szczecin , Poland
| | - Magdalena Nalewajska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University , Szczecin , Poland
| | | | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University , Szczecin , Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University , Szczecin , Poland
| |
Collapse
|
18
|
Césaire M, Thariat J, Candéias SM, Stefan D, Saintigny Y, Chevalier F. Combining PARP inhibition, radiation, and immunotherapy: A possible strategy to improve the treatment of cancer? Int J Mol Sci 2018; 19:ijms19123793. [PMID: 30487462 PMCID: PMC6321381 DOI: 10.3390/ijms19123793] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/20/2018] [Accepted: 11/24/2018] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy has revolutionized the practice of oncology, improving survival in certain groups of patients with cancer. Immunotherapy can synergize with radiation therapy, increase locoregional control, and have abscopal effects. Combining it with other treatments, such as targeted therapies, is a promising means of improving the efficacy of immunotherapy. Because the value of immunotherapy is amplified with the expression of tumor antigens, coupling poly(ADP-ribose) polymerase (PARP) inhibitors and immunotherapy might be a promising treatment for cancer. Further, PARP inhibitors (PARPis) are being combined with radiation therapy to inhibit DNA repair functions, thus enhancing the effects of radiation; this association might interact with the antitumor immune response. Cytotoxic T lymphocytes are central to the antitumor immune response. PARP inhibitors and ionizing radiation can enhance the infiltration of cytotoxic T lymphocytes into the tumor bed, but they can also enhance PD-1/PDL-1 expression. Thus, the addition of immune checkpoint inhibitors with PARP inhibitors and/or ionizing radiation could counterbalance such immunosuppressive effects. With the present review article, we proposed to evaluate some of these associated therapies, and we explored the biological mechanisms and medical benefits of the potential combination of radiation therapy, immunotherapy, and PARP inhibitors.
Collapse
Affiliation(s)
- Mathieu Césaire
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14076 Caen, France.
- UMR6252 CIMAP, CEA - CNRS - ENSICAEN - Université de Caen Normandie, 14076 Caen, France.
- Radiotherapy Unit, Centre François Baclesse, 14000 Caen, France.
| | - Juliette Thariat
- Radiotherapy Unit, Centre François Baclesse, 14000 Caen, France.
| | - Serge M Candéias
- ProMD, Chemistry and Biology of Metals Laboratory, Univ. Grenoble Alpes, CEA, CNRS, BIG-LCBM, 38054 Grenoble, France.
| | - Dinu Stefan
- Radiotherapy Unit, Centre François Baclesse, 14000 Caen, France.
| | - Yannick Saintigny
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14076 Caen, France.
- UMR6252 CIMAP, CEA - CNRS - ENSICAEN - Université de Caen Normandie, 14076 Caen, France.
| | - François Chevalier
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14076 Caen, France.
- UMR6252 CIMAP, CEA - CNRS - ENSICAEN - Université de Caen Normandie, 14076 Caen, France.
| |
Collapse
|
19
|
ABT-888 restores sensitivity in temozolomide resistant glioma cells and xenografts. PLoS One 2018; 13:e0202860. [PMID: 30153289 PMCID: PMC6112648 DOI: 10.1371/journal.pone.0202860] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/12/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Temozolomide (TMZ) is active against glioblastomas (GBM) in which the O6-methylguanine-DNA methyltransferase (MGMT) gene is silenced. However, even in responsive cases, its beneficial effect is undermined by the emergence of drug resistance. Here, we tested whether inhibition of poly (ADP-ribose) polymerase-1 and -2 (PARP) enhanced the effectiveness of TMZ. METHODS Using patient derived brain tumor initiating cells (BTICs) and orthotopic xenografts as models of newly diagnosed and recurrent high-grade glioma, we assessed the effects of TMZ, ABT-888, and the combination of TMZ and ABT-888 on the viability of BTICs and survival of tumor-bearing mice. We also studied DNA damage repair, checkpoint protein phosphorylation, and DNA replication in mismatch repair (MMR) deficient cells treated with TMZ and TMZ plus ABT-888. RESULTS Cells and xenografts derived from newly diagnosed MGMT methylated high-grade gliomas were sensitive to TMZ while those derived from unmethylated and recurrent gliomas were typically resistant. ABT-888 had no effect on the viability of BTICs or tumor bearing mice, but co-treatment with TMZ restored sensitivity in resistant cells and xenografts from newly diagnosed unmethylated gliomas and recurrent gliomas with MSH6 mutations. In contrast, the addition of ABT-888 to TMZ had little sensitizing effect on cells and xenografts derived from newly diagnosed methylated gliomas. In a model of acquired TMZ resistance mediated by loss of MMR gene MSH6, re-sensitization to TMZ by ABT-888 was accompanied by persistent DNA strand breaks, re-engagement of checkpoint kinase signaling, and interruption of DNA synthesis. CONCLUSION In laboratory models, the addition of ABT-888 to TMZ overcame resistance to TMZ.
Collapse
|
20
|
Understanding Resistance Mechanisms and Expanding the Therapeutic Utility of PARP Inhibitors. Cancers (Basel) 2017; 9:cancers9080109. [PMID: 28829366 PMCID: PMC5575612 DOI: 10.3390/cancers9080109] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 11/20/2022] Open
Abstract
Poly-(ADP-ribose) polymerase (PARP) inhibitors act through synthetic lethality in cells with defects in homologous recombination (HR) DNA repair caused by molecular aberrations such as BRCA mutations, and is approved for treatment in ovarian cancer, with promising clinical activity against other HR defective tumors including breast and prostate cancers. Three PARP inhibitors have been FDA approved, while another two have shown promising activity and are in late stage development. Nonetheless, both primary and secondary resistance to PARP inhibition have led to treatment failure, and the development of predictive biomarkers and the ability to identify and overcome mechanisms of resistance is vital for optimization of its clinical utility. Additionally, there has been evidence that PARP inhibition may have a therapeutic role beyond HR deficient tumors which warrants further investigation, both as single agent and in combination with other therapeutic modalities like cytotoxic chemotherapy, radiation, targeted therapy and immunotherapy. With new strategies to overcome resistance and expand its therapeutic utility, PARP inhibitors are likely to become a staple in our armamentarium of drugs in cancer therapeutics.
Collapse
|
21
|
Castle KD, Chen M, Wisdom AJ, Kirsch DG. Genetically engineered mouse models for studying radiation biology. Transl Cancer Res 2017; 6:S900-S913. [PMID: 30733931 PMCID: PMC6363345 DOI: 10.21037/tcr.2017.06.19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetically engineered mouse models (GEMMs) are valuable research tools that have transformed our understanding of cancer. The first GEMMs generated in the 1980s and 1990s were knock-in and knock-out models of single oncogenes or tumor suppressors. The advances that made these models possible catalyzed both technological and conceptual shifts in the way cancer research was conducted. As a result, dozens of mouse models of cancer exist today, covering nearly every tissue type. The advantages inherent to GEMMs compared to in vitro and in vivo transplant models are compounded in preclinical radiobiology research for several reasons. First, they accurately and robustly recapitulate primary cancers anatomically, histopathologically, and genetically. Reliable models are a prerequisite for predictive preclinical studies. Second, they preserve the tumor microenvironment, including the immune, vascular, and stromal compartments, which enables the study of radiobiology at a systems biology level. Third, they provide exquisite control over the genetics and kinetics of tumor initiation, which enables the study of specific gene mutations on radiation response and functional genomics in vivo. Taken together, these facets allow researchers to utilize GEMMs for rigorous and reproducible preclinical research. In the three decades since the generation of the first GEMMs of cancer, advancements in modeling approaches have rapidly progressed and expanded the mouse modeling toolbox with techniques such as in vivo short hairpin RNA (shRNA) knockdown, inducible gene expression, site-specific recombinases, and dual recombinase systems. Our lab and many others have utilized these tools to study cancer and radiobiology. Recent advances in genome engineering with CRISPR/Cas9 technology have made GEMMs even more accessible to researchers. Here, we review current and future approaches to mouse modeling with a focus on applications in preclinical radiobiology research.
Collapse
Affiliation(s)
- Katherine D. Castle
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mark Chen
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA
| | - Amy J. Wisdom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA
| | - David G. Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
22
|
Jue TR, Nozue K, Lester AJ, Joshi S, Schroder LBW, Whittaker SP, Nixdorf S, Rapkins RW, Khasraw M, McDonald KL. Veliparib in combination with radiotherapy for the treatment of MGMT unmethylated glioblastoma. J Transl Med 2017; 15:61. [PMID: 28314386 PMCID: PMC5356284 DOI: 10.1186/s12967-017-1164-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/13/2017] [Indexed: 12/16/2022] Open
Abstract
Background The O6-methylguanine methyltransferase (MGMT) gene is frequently unmethylated in patients with glioblastoma (GBM), rendering them non-responsive to the standard treatment regime of surgery followed by concurrent radiotherapy (RT) and temozolomide. Here, we investigate the efficacy of adding a PARP inhibitor, veliparib, to radiotherapy to treat MGMT unmethylated GBM. Methods The inhibition of PARP with veliparib (ABT-888), a potent and orally bioavailable inhibitor in combination with RT was tested on a panel of patient derived cell lines (PDCLs) and patient-derived xenografts (PDX) models generated from GBM patients with MGMT unmethylated tumors. Results The combination of veliparib and RT inhibited colony formation in the majority of PDCLs tested. The PDCL, RN1 showed significantly reduced levels of the homologous repair protein, Mre11 and a heightened response to PARP inhibition measured by increased apoptosis and decreased colony formation. The oral administration of veliparib (12.5 mg/kg, twice daily for 5 days in a 28-day treatment cycle) in combination with whole brain RT (4 Gy) induced apoptosis (Tunel staining) and decreased cell proliferation (Ki67 staining) in a PDX of MGMT unmethylated GBM. Significantly longer survival times of the PDX treated with the combination treatment were recorded compared to RT only or veliparib only. Conclusions Our results demonstrate preclinical efficacy of targeting PARP at multiple levels and provide a new approach for the treatment of MGMT unmethylated GBM.
Collapse
Affiliation(s)
- Toni Rose Jue
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Kyoko Nozue
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Ashleigh J Lester
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Swapna Joshi
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Lisette B W Schroder
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Shane P Whittaker
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Sheri Nixdorf
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | - Robert W Rapkins
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia
| | | | - Kerrie L McDonald
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, Adult Cancer Program, Lowy Cancer Research Centre, UNSW, Kensington, Australia.
| |
Collapse
|
23
|
Somnay Y, Lubner S, Gill H, Matsumura JB, Chen H. The PARP inhibitor ABT-888 potentiates dacarbazine-induced cell death in carcinoids. Cancer Gene Ther 2016; 23:348-354. [PMID: 27632933 PMCID: PMC5083201 DOI: 10.1038/cgt.2016.39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023]
Abstract
Monoagent DNA-alkylating chemotherapies like dacarbazine are among a paucity of medical treatments for advanced carcinoid tumors, but are limited by host toxicity and intrinsic chemoresistance through the base excision repair (BER) pathway via poly (ADP-ribose) polymerase (PARP). Hence, inhibitors of PARP may potentiate DNA-damaging agents by blocking BER and DNA restoration. We show that the PARP inhibitor ABT-888 (Veliparib) enhances the cytotoxic effects of dacarbazine in carcinoids. Two human carcinoid cell lines (BON and H727) treated with a combination of ABT-888 and dacarbazine resulted in synergistic growth inhibition signified by combination indices <1 on the Chou-Talalay scale. ABT-888 administered prior to varying dacarbazine doses promoted the suppression of neuroendocrine biomarkers of malignancy ASCL1 and CgA, shown by Western analysis. ATM phosphorylation and p21Waf1/Cip1 activation, indicative of DNA damage, were increased by ABT-888 when combined with dacarbazine treatment, suggesting BER pathway attenuation by ABT-888. PE Annexin V/7-AAD staining and sorting revealed a profound induction of apoptosis following combination treatment, which was further confirmed by increased PARP cleavage. These results demonstrate that ABT-888 synergizes dacarbazine treatment in carcinoids. Therefore, ABT-888 may help treat carcinoids unresponsive or refractory to mainstay therapies.
Collapse
Affiliation(s)
- Y Somnay
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - S Lubner
- Division of Hematology and Medical Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - H Gill
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J B Matsumura
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - H Chen
- Endocrine Surgery Research Laboratories, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Surgery, University of Alabama- Birmingham, Birmingham, AL, USA
| |
Collapse
|