DNA Repair Inhibitors: Potential Targets and Partners for Targeted Radionuclide Therapy

The present review aims to explore the potential targets/partners for future targeted radionuclide therapy (TRT) strategies, wherein cancer cells often are not killed effectively, despite receiving a high average tumor radiation dose. Here, we shall discuss the key factors in the cancer genome, especially those related to DNA damage response/repair and maintenance systems for escaping cell death in cancer cells. To overcome the current limitations of TRT effectiveness due to radiation/drug-tolerant cells and tumor heterogeneity, and to make TRT more effective, we propose that a promising strategy would be to target the DNA maintenance factors that are crucial for cancer survival. Considering their cancer-specific DNA damage response/repair ability and dysregulated transcription/epigenetic system, key factors such as PARP, ATM/ATR, amplified/overexpressed transcription factors, and DNA methyltransferases have the potential to be molecular targets for Auger electron therapy; moreover, their inhibition by non-radioactive molecules could be a partnering component for enhancing the therapeutic response of TRT.

[1]  M. Deng,et al.  DNA Repair Deficiency Regulates Immunity Response in Cancers: Molecular Mechanism and Approaches for Combining Immunotherapy , 2023, Cancers.

[2]  P. Albertsson,et al.  Astatine-211 based radionuclide therapy: Current clinical trial landscape , 2023, Frontiers in Medicine.

[3]  B. Cornelissen,et al.  A radioiodinated rucaparib analogue as an Auger electron emitter for cancer therapy. , 2022, Nuclear medicine and biology.

[4]  F. Mottaghy,et al.  PARP targeted Auger emitter therapy with [125I]PARPi-01 for triple-negative breast cancer , 2022, EJNMMI Research.

[5]  Xu Liu,et al.  Radiotherapy combined with immunotherapy: the dawn of cancer treatment , 2022, Signal Transduction and Targeted Therapy.

[6]  Hui Xu,et al.  A New Trend in Cancer Treatment: The Combination of Epigenetics and Immunotherapy , 2022, Frontiers in Immunology.

[7]  F. Mottaghy,et al.  Auger Emitter Conjugated PARP Inhibitor for Therapy in Triple Negative Breast Cancers: A Comparative In-Vitro Study , 2022, Cancers.

[8]  E. Mittra,et al.  177Lu-Dotatate plus long-acting octreotide versus high‑dose long-acting octreotide in patients with midgut neuroendocrine tumours (NETTER-1): final overall survival and long-term safety results from an open-label, randomised, controlled, phase 3 trial. , 2021, The Lancet. Oncology.

[9]  M. Baretti,et al.  Epigenetic modifiers synergize with immune-checkpoint blockade to enhance long-lasting antitumor efficacy. , 2021, The Journal of clinical investigation.

[10]  C. Lord,et al.  Targeting the DNA damage response in immuno-oncology: developments and opportunities , 2021, Nature Reviews Cancer.

[11]  T. Reiner,et al.  PARP-Targeted Auger Therapy in p53 Mutant Colon Cancer Xenograft Mouse Models. , 2021, Molecular pharmaceutics.

[12]  M. He,et al.  Molecular mechanisms of chemo‐ and radiotherapy resistance and the potential implications for cancer treatment , 2021, MedComm.

[13]  F. Haddad,et al.  Overview of the Most Promising Radionuclides for Targeted Alpha Therapy: The “Hopeful Eight” , 2021, Pharmaceutics.

[14]  S. Rottenberg,et al.  Studying PAR-Dependent Chromatin Remodeling to Tackle PARPi Resistance. , 2021, Trends in molecular medicine.

[15]  J. Licht,et al.  Leveraging epigenetics to enhance the efficacy of immunotherapy , 2021, Clinical Epigenetics.

[16]  Jacob D. Jaffe,et al.  Epigenetic silencing by SETDB1 suppresses tumour intrinsic immunogenicity , 2021, Nature.

[17]  J. Liou,et al.  Recent developments in epigenetic cancer therapeutics: clinical advancement and emerging trends , 2021, Journal of Biomedical Science.

[18]  R. Mach,et al.  PARP Targeted Alpha-Particle Therapy Enhances Response to PD-1 Immune-Checkpoint Blockade in a Syngeneic Mouse Model of Glioblastoma. , 2021, ACS pharmacology & translational science.

[19]  Qiulian Wu,et al.  FBXO44 promotes DNA replication-coupled repetitive element silencing in cancer cells , 2020, Cell.

[20]  J. Carles,et al.  Advanced Prostate Cancer with ATM Loss: PARP and ATR Inhibitors. , 2020, European urology.

[21]  Y. Drew,et al.  First-in-Human Trial of the Oral Ataxia Telangiectasia and Rad3-Related Inhibitor BAY 1895344 in Patients with Advanced Solid Tumors. , 2020, Cancer discovery.

[22]  M. McDevitt,et al.  Radiopharmaceutical therapy in cancer: clinical advances and challenges , 2020, Nature Reviews Drug Discovery.

[23]  N. Magné,et al.  Radiation-induced bystander and abscopal effects: important lessons from preclinical models , 2020, British Journal of Cancer.

[24]  R. Tothill,et al.  Enhancing the anti-tumour activity of 177Lu-DOTA-octreotate radionuclide therapy in somatostatin receptor-2 expressing tumour models by targeting PARP , 2020, Scientific Reports.

[25]  E. Sala,et al.  The emerging role of cell surface receptor and protein binding radiopharmaceuticals in cancer diagnostics and therapy. , 2020, Nuclear medicine and biology.

[26]  P. Zhou,et al.  DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer , 2020, Signal Transduction and Targeted Therapy.

[27]  W. Hahn,et al.  ATM Loss Confers Greater Sensitivity to ATR Inhibition Than PARP Inhibition in Prostate Cancer , 2020, Cancer Research.

[28]  D. Slade PARP and PARG inhibitors in cancer treatment. , 2020, Genes & development.

[29]  Kosei Ito,et al.  Oncogenic RUNX3: A Link between p53 Deficiency and MYC Dysregulation , 2020, Molecules and cells.

[30]  S. Ghasemi Cancer's epigenetic drugs: where are they in the cancer medicines? , 2019, The Pharmacogenomics Journal.

[31]  Junhong Han,et al.  Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials , 2019, Signal Transduction and Targeted Therapy.

[32]  A. S. Sobolev,et al.  Delivery systems exploiting natural cell transport processes of macromolecules for intracellular targeting of Auger electron emitters. , 2019, Nuclear medicine and biology.

[33]  R. Mach,et al.  PARP-1–Targeted Auger Emitters Display High-LET Cytotoxic Properties In Vitro but Show Limited Therapeutic Utility in Solid Tumor Models of Human Neuroblastoma , 2019, The Journal of Nuclear Medicine.

[34]  H. Bahig,et al.  Spatially fractionated radiation therapy: History, present and the future , 2019, Clinical and translational radiation oncology.

[35]  R. Reilly,et al.  Auger electrons for cancer therapy – a review , 2019, EJNMMI Radiopharmacy and Chemistry.

[36]  Jung Kyoon Choi,et al.  DNA methylation loss promotes immune evasion of tumours with high mutation and copy number load , 2019, Nature Communications.

[37]  J. Bushweller Targeting transcription factors in cancer — from undruggable to reality , 2019, Nature Reviews Cancer.

[38]  Reshma Jagsi,et al.  PARP1 Inhibition Radiosensitizes Models of Inflammatory Breast Cancer to Ionizing Radiation , 2019, Molecular Cancer Therapeutics.

[39]  Jie Huang,et al.  Upregulation of RUNX1 Suppresses Proliferation and Migration through Repressing VEGFA Expression in Hepatocellular Carcinoma , 2019, Pathology & Oncology Research.

[40]  Andrew J. Bannister,et al.  The roles of DNA, RNA and histone methylation in ageing and cancer , 2019, Nature Reviews Molecular Cell Biology.

[41]  H. Sugiyama,et al.  Therapeutic gene regulation using pyrrole-imidazole polyamides. , 2019, Advanced drug delivery reviews.

[42]  J. Humm,et al.  Targeted Brain Tumor Radiotherapy Using an Auger Emitter , 2019, Clinical Cancer Research.

[43]  R. Mach,et al.  Targeting PARP-1 with Alpha-Particles Is Potently Cytotoxic to Human Neuroblastoma in Preclinical Models , 2019, Molecular Cancer Therapeutics.

[44]  C. Liang,et al.  N-Myc promotes therapeutic resistance development of neuroendocrine prostate cancer by differentially regulating miR-421/ATM pathway , 2019, Molecular Cancer.

[45]  A. S. Sobolev,et al.  Antitumor Activity of Auger Electron Emitter 111In Delivered by Modular Nanotransporter for Treatment of Bladder Cancer With EGFR Overexpression , 2018, Front. Pharmacol..

[46]  M. Esteller,et al.  Clinical epigenetics: seizing opportunities for translation , 2018, Nature Reviews Genetics.

[47]  T. Thompson,et al.  N-MYC regulation of DNA damage response in neuroendocrine prostate cancer: mechanistic insight and novel combination therapy approaches , 2018, Oncoscience.

[48]  S. Demaria,et al.  Immunological Mechanisms Responsible for Radiation-Induced Abscopal Effect. , 2018, Trends in immunology.

[49]  E. Lecona,et al.  Targeting ATR in cancer , 2018, Nature Reviews Cancer.

[50]  R. Young,et al.  Selective gene dependencies in MYCN-amplified neuroblastoma include the core transcriptional regulatory circuitry , 2018, Nature Genetics.

[51]  Y. Kaneda,et al.  RUNX1 positively regulates the ErbB2/HER2 signaling pathway through modulating SOS1 expression in gastric cancer cells , 2018, Scientific Reports.

[52]  Z. Zeng,et al.  Role of metabolism in cancer cell radioresistance and radiosensitization methods , 2018, Journal of experimental & clinical cancer research : CR.

[53]  J. Hesser,et al.  Using immunotherapy to boost the abscopal effect , 2018, Nature Reviews Cancer.

[54]  M. Lieber,et al.  Nonhomologous DNA end-joining for repair of DNA double-strand breaks , 2017, The Journal of Biological Chemistry.

[55]  A. Chinnaiyan,et al.  Targeting the MYCN–PARP–DNA Damage Response Pathway in Neuroendocrine Prostate Cancer , 2017, Clinical Cancer Research.

[56]  Timothy W. Secomb,et al.  Transport of drugs from blood vessels to tumour tissue , 2017, Nature Reviews Cancer.

[57]  J. Habrand,et al.  Poly-(ADP-ribose)-polymerase inhibitors as radiosensitizers: a systematic review of pre-clinical and clinical human studies , 2017, Oncotarget.

[58]  B. Arumugam,et al.  Role of Runx2 in breast cancer-mediated bone metastasis. , 2017, International journal of biological macromolecules.

[59]  A. Mai,et al.  Epi-drugs in combination with immunotherapy: a new avenue to improve anticancer efficacy , 2017, Clinical Epigenetics.

[60]  Paolo Fortina,et al.  Transcriptomic profiling of 39 commonly-used neuroblastoma cell lines , 2017, Scientific Data.

[61]  Alan Ashworth,et al.  PARP inhibitors: Synthetic lethality in the clinic , 2017, Science.

[62]  P. Vaz,et al.  Evaluation of Acridine Orange Derivatives as DNA-Targeted Radiopharmaceuticals for Auger Therapy: Influence of the Radionuclide and Distance to DNA , 2017, Scientific Reports.

[63]  A. Nussenzweig,et al.  Endogenous DNA Damage as a Source of Genomic Instability in Cancer , 2017, Cell.

[64]  R. Young,et al.  Transcriptional Addiction in Cancer , 2017, Cell.

[65]  Haitao Zhao,et al.  Mps1/TTK: a novel target and biomarker for cancer , 2017, Journal of drug targeting.

[66]  Julio Caballero,et al.  Direct and Auger Electron-Induced, Single- and Double-Strand Breaks on Plasmid DNA Caused by 99mTc-Labeled Pyrene Derivatives and the Effect of Bonding Distance , 2016, PloS one.

[67]  R. Mach,et al.  A Radiotracer Strategy to Quantify PARP-1 Expression In Vivo Provides a Biomarker That Can Enable Patient Selection for PARP Inhibitor Therapy. , 2016, Cancer research.

[68]  T. Kipps,et al.  ATM Mutations in Cancer: Therapeutic Implications , 2016, Molecular Cancer Therapeutics.

[69]  F. Mottaghy,et al.  225Ac-PSMA-617 for PSMA-Targeted α-Radiation Therapy of Metastatic Castration-Resistant Prostate Cancer , 2016, The Journal of Nuclear Medicine.

[70]  C. Allis,et al.  The molecular hallmarks of epigenetic control , 2016, Nature Reviews Genetics.

[71]  T. Mak,et al.  Targeting Mitosis in Cancer: Emerging Strategies. , 2015, Molecular cell.

[72]  R. Medema,et al.  Inhibition of the spindle assembly checkpoint kinase TTK enhances the efficacy of docetaxel in a triple-negative breast cancer model. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[73]  T. Paull Mechanisms of ATM Activation. , 2015, Annual review of biochemistry.

[74]  Kosuke Saito,et al.  Preclinical Study of Novel Gene Silencer Pyrrole-Imidazole Polyamide Targeting Human TGF-β1 Promoter for Hypertrophic Scars in a Common Marmoset Primate Model , 2015, PloS one.

[75]  H. Ishwaran,et al.  Radiation and Dual Checkpoint Blockade Activates Non-Redundant Immune Mechanisms in Cancer , 2015, Nature.

[76]  Yoshiaki Ito,et al.  The RUNX family: developmental regulators in cancer , 2015, Nature Reviews Cancer.

[77]  K. Knudsen,et al.  Beyond DNA repair: DNA-PK function in cancer. , 2014, Cancer discovery.

[78]  Phillip A. Sharp,et al.  Target specificity of the CRISPR-Cas9 system , 2014, Quantitative Biology.

[79]  Joanna S Morris,et al.  Expression of RUNX1 Correlates with Poor Patient Prognosis in Triple Negative Breast Cancer , 2014, PloS one.

[80]  S. Lees-Miller,et al.  Low ATM protein expression and depletion of p53 correlates with olaparib sensitivity in gastric cancer cell lines , 2014, Cell cycle.

[81]  David J. Chen,et al.  DNA-PK: a dynamic enzyme in a versatile DSB repair pathway. , 2014, DNA repair.

[82]  G. Lockwood,et al.  Phase I trial to evaluate the tumor and normal tissue uptake, radiation dosimetry and safety of (111)In-DTPA-human epidermal growth factor in patients with metastatic EGFR-positive breast cancer. , 2014, American journal of nuclear medicine and molecular imaging.

[83]  M. Gaze,et al.  A systematic review of 131I-meta iodobenzylguanidine molecular radiotherapy for neuroblastoma. , 2014, European journal of cancer.

[84]  P. Stephens,et al.  Radium Ra 223 Dichloride Injection: U.S. Food and Drug Administration Drug Approval Summary , 2013, Clinical Cancer Research.

[85]  William A Weiss,et al.  Neuroblastoma and MYCN. , 2013, Cold Spring Harbor perspectives in medicine.

[86]  M. Lomax,et al.  Biological consequences of radiation-induced DNA damage: relevance to radiotherapy. , 2013, Clinical oncology (Royal College of Radiologists (Great Britain)).

[87]  B. Kaina,et al.  Contribution of ATM and ATR to the Resistance of Glioblastoma and Malignant Melanoma Cells to the Methylating Anticancer Drug Temozolomide , 2013, Molecular Cancer Therapeutics.

[88]  E. Yang,et al.  PARP-1: Friend or Foe of DNA Damage and Repair in Tumorigenesis? , 2013, Cancers.

[89]  David E. Muench,et al.  c-Myc and Cancer Metabolism , 2012, Clinical Cancer Research.

[90]  Guido Marcucci,et al.  RUNX1 mutations are associated with poor outcome in younger and older patients with cytogenetically normal acute myeloid leukemia and with distinct gene and MicroRNA expression signatures. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[91]  Xiang Xu,et al.  Effect of distance between decaying 125I and DNA on Auger-electron induced double-strand break yield , 2012, International journal of radiation biology.

[92]  O. Barakat,et al.  Long-Term Survival, Toxicity Profile, and role of F-18 FDG PET/CT scan in Patients with Progressive Neuroendocrine Tumors Following Peptide Receptor Radionuclide Therapy with High Activity In-111 Pentetreotide , 2012, Theranostics.

[93]  Michael Jones,et al.  Intragenic ATM methylation in peripheral blood DNA as a biomarker of breast cancer risk. , 2012, Cancer research.

[94]  B. Telfer,et al.  Inhibition of PARP-1 by Olaparib (AZD2281) Increases the Radiosensitivity of a Lung Tumor Xenograft , 2011, Molecular Cancer Therapeutics.

[95]  G. Stein,et al.  The Role of RUNX2 in Osteosarcoma Oncogenesis , 2010, Sarcoma.

[96]  A. Shibata,et al.  Sensitization to Radiation and Alkylating Agents by Inhibitors of Poly(ADP-ribose) Polymerase Is Enhanced in Cells Deficient in DNA Double-Strand Break Repair , 2010, Molecular Cancer Therapeutics.

[97]  C. Bascoul-Mollevi,et al.  Author manuscript, published in "Journal of nuclear medicine: official publication, Society of Nuclear Medicine 2009;50 (12):2033-41" , 2010 .

[98]  N. Kawashima,et al.  Fractionated but Not Single-Dose Radiotherapy Induces an Immune-Mediated Abscopal Effect when Combined with Anti–CTLA-4 Antibody , 2009, Clinical Cancer Research.

[99]  Vivian G. Cheung,et al.  Genetic analysis of radiation-induced changes in human gene expression , 2009, Nature.

[100]  Kevin M. Prise,et al.  Radiation-induced bystander signalling in cancer therapy , 2009, Nature Reviews Cancer.

[101]  J. Peto,et al.  Gene-body hypermethylation of ATM in peripheral blood DNA of bilateral breast cancer patients , 2009, Human molecular genetics.

[102]  A. Chalmers,et al.  Replication-dependent radiosensitization of human glioma cells by inhibition of poly(ADP-Ribose) polymerase: mechanisms and therapeutic potential. , 2008, International journal of radiation oncology, biology, physics.

[103]  M. Spitz,et al.  Prognostic significance of ataxia‐telangiectasia mutated, DNA‐dependent protein kinase catalytic subunit, and Ku heterodimeric regulatory complex 86‐kD subunit expression in patients with nonsmall cell lung cancer , 2008, Cancer.

[104]  S. Adelstein,et al.  First human treatment of resistant neoplastic meningitis by intrathecal administration of MTX Plus 125IUdR , 2008, International journal of radiation biology.

[105]  D. Park,et al.  Altered expression of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) during gastric carcinogenesis and its clinical implications on gastric cancer. , 2007, International journal of oncology.

[106]  T. Karagiannis Comparison of different classes of radionuclides for potential use in radioimmunotherapy. , 2007, Hellenic journal of nuclear medicine.

[107]  P. Fisher,et al.  Radiation-induced cell signaling: inside-out and outside-in , 2007, Molecular Cancer Therapeutics.

[108]  G. Stein,et al.  Regulatory roles of Runx2 in metastatic tumor and cancer cell interactions with bone , 2006, Cancer and Metastasis Reviews.

[109]  D. Chung,et al.  MYCN silencing induces differentiation and apoptosis in human neuroblastoma cells. , 2006, Biochemical and biophysical research communications.

[110]  S. Tanada,et al.  Molecular therapy of human neuroblastoma cells using Auger electrons of 111In-labeled N-myc antisense oligonucleotides. , 2006, Journal of nuclear medicine : official publication, Society of Nuclear Medicine.

[111]  Franz Buchegger,et al.  Auger radiation targeted into DNA: a therapy perspective , 2006, European Journal of Nuclear Medicine and Molecular Imaging.

[112]  C. Godon,et al.  Radiosensitization by the poly(ADP-ribose) polymerase inhibitor 4-amino-1,8-naphthalimide is specific of the S phase of the cell cycle and involves arrest of DNA synthesis , 2006, Molecular Cancer Therapeutics.

[113]  S. Baylin,et al.  Epigenetic gene silencing in cancer – a mechanism for early oncogenic pathway addiction? , 2006, Nature Reviews Cancer.

[114]  Lesley McGuffog,et al.  Cancer risks and mortality in heterozygous ATM mutation carriers. , 2005, Journal of the National Cancer Institute.

[115]  Thomas Helleday,et al.  Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase , 2005, Nature.

[116]  Alan Ashworth,et al.  Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy , 2005, Nature.

[117]  S. Lees-Miller,et al.  DNA damage-induced activation of ATM and ATM-dependent signaling pathways. , 2004, DNA repair.

[118]  Peter A. Jones,et al.  Epigenetics in human disease and prospects for epigenetic therapy , 2004, Nature.

[119]  Luther W Brady,et al.  Radioimmunotherapy as a novel treatment regimen: 125I-labeled monoclonal antibody 425 in the treatment of high-grade brain gliomas. , 2004, International journal of radiation oncology, biology, physics.

[120]  Kai Rothkamm,et al.  A Double-Strand Break Repair Defect in ATM-Deficient Cells Contributes to Radiosensitivity , 2004, Cancer Research.

[121]  K. Brown,et al.  Ataxia-Telangiectasia-Mutated (ATM) Gene in Head and Neck Squamous Cell Carcinoma , 2004, Cancer Epidemiology Biomarkers & Prevention.

[122]  L. Gordon,et al.  Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin's lymphoma. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[123]  J. Griffith,et al.  Preferential binding of ATR protein to UV-damaged DNA , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[124]  L. Kvols,et al.  Somatostatin receptor-targeted radionuclide therapy of tumors: preclinical and clinical findings. , 2002, Seminars in nuclear medicine.

[125]  L. Marnett,et al.  Endogenous DNA damage and mutation. , 2001, Trends in genetics : TIG.

[126]  P. Schär,et al.  Spontaneous DNA Damage, Genome Instability, and Cancer—When DNA Replication Escapes Control , 2001, Cell.

[127]  B. Hann,et al.  Purification and DNA binding properties of the ataxia-telangiectasia gene product ATM. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[128]  G. Poirier,et al.  Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. , 1999, The Biochemical journal.

[129]  X. Le,et al.  Inducible repair of thymine glycol detected by an ultrasensitive assay for DNA damage. , 1998, Science.

[130]  D. Fuster,et al.  Strontium-89 for palliation of pain from bone metastases in patients with prostate and breast cancer , 1997, European Journal of Nuclear Medicine.

[131]  J Isola,et al.  Androgen receptor gene amplification: a possible molecular mechanism for androgen deprivation therapy failure in prostate cancer. , 1997, Cancer research.

[132]  L. Hartwell,et al.  Cell cycle control and cancer. , 1994, Science.

[133]  A. Scott,et al.  Phase I/II study of iodine 125-labeled monoclonal antibody A33 in patients with advanced colon cancer. , 1994, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[134]  Peter A. Jones,et al.  Epigenetics in cancer. , 2010, Carcinogenesis.

[135]  S. Adelstein,et al.  Radiobiologic principles in radionuclide therapy. , 2005, Journal of nuclear medicine : official publication, Society of Nuclear Medicine.

[136]  J. Cadet,et al.  Measurement of DNA base damage in cells exposed to low doses of gamma-radiation: comparison between the HPLC-EC and comet assays. , 1999, International journal of radiation biology.

[137]  J. Ward,et al.  DNA damage produced by ionizing radiation in mammalian cells: identities, mechanisms of formation, and reparability. , 1988, Progress in nucleic acid research and molecular biology.