122
Views
0
CrossRef citations to date
0
Altmetric
Review

Unblinding the watchmaker: cancer treatment and drug design in the face of evolutionary pressure

, , , , , , , , , & show all
Pages 1081-1094 | Received 20 Feb 2022, Accepted 15 Aug 2022, Published online: 30 Aug 2022

References

  • Kattner P, Zeiler K, Herbener VJ, et al. What animal cancers teach us about human biology. Theranostics. 2021;11:6682–6702.
  • Tomasetti C, Vogelstein B. Cancer etiology. variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science. 2015;347:78–81.
  • Wodarz D, Zauber AG. Cancer: risk factors and random chances. Nature. 2015;517:563–564.
  • Weinberg CR, Zaykin D. Is bad luck the main cause of cancer? J Natl Cancer Inst. 2015;107:djv125–djv125.
  • Rozhok AI, Wahl GM, DeGregori J. A critical examination of the “bad luck” explanation of cancer risk. Cancer Prev Res (Phila). 2015;8:762–764.
  • Wu S, Powers S, Zhu W, et al. Substantial contribution of extrinsic risk factors to cancer development. Nature. 2016;529:43–47.
  • White MC, Holman DM, Boehm JE, et al. Age and cancer risk: a potentially modifiable relationship. Am J Prev Med. 2014;46:7.
  • Dunn GP, Old LJ, Schreiber RD. The three es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–360.
  • Ohgaki H, Kleihues P. The definition of primary and secondary glioblastoma. Clin Cancer Res. 2013;19:764–772.
  • Srivastava S, Ghosh S,J, Kagan J, et al. The making of a precancer atlas: promises, challenges, and opportunities. Trends Cancer. 2018;4:523–536.
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674.
  • Esmatabadi MJD, Bakhshinejad B, Motlagh FM, et al. Therapeutic resistance and cancer recurrence mechanisms: unfolding the story of tumour coming back. J Biosci. 2016;41:497–506.
  • Nikolaou M, Pavlopoulou A, Georgakilas AG, et al. The challenge of drug resistance in cancer treatment: a current overview. Clin Exp Metastasis. 2018;35:309–318.
  • Chatterjee N, Bivona TG. Polytherapy and targeted cancer drug resistance. Trends Cancer. 2019;5:170–182.
  • Goldie JH. Drug resistance in cancer: a perspective. Cancer Metastasis Rev. 2001;20:63–68.
  • Nonnenmacher L, Hasslacher S, Zimmermann J, et al. Cell death induction in cancer therapy - past, present, and future. Crit Rev Oncog. 2016;21:253–267.
  • Li L, Guan Y, Chen X, et al. DNA repair pathways in cancer therapy and resistance. Front Pharmacol. 2020;11:629266.
  • Robey RW, Pluchino RM, Hall MD, et al. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer. 2018;18:452–464.
  • Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019;2:141–160.
  • Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–998.
  • Ribatti D. The concept of immune surveillance against tumors: the first theories. Oncotarget. 2017;8:7175–7180.
  • Fares J, Fares MY, Khachfe HH, et al. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct Target Ther. 2020;5.
  • Luzzi KJ, MacDonald IC, Schmidt EE, et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. The American Journal of Pathology. 1998;153:865–873.
  • Wang W-C, Zhang X-F, Peng J, et al. Survival mechanisms and influence factors of circulating tumor cells. BioMed Research International. 2018;2018:6304701.
  • Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nature Medicine. 2006;12:895–904.
  • López-Lázaro M. Why do tumors metastasize? Cancer Biology & Therapy. 2007;6:141–144.
  • Tomlinson I, Sasieni P, Bodmer W. How many mutations in a cancer? The American Journal of Pathology. 2002;160:755–758.
  • Riviere P, Goodman AM, Okamura R, et al. High tumor mutational burden correlates with longer survival in immunotherapy-naïve patients with diverse cancers. Molecular Cancer Therapeutics. 2020;19:2139–2145.
  • Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Medicine. 2017;9:34.
  • Westhoff M-A, Marschall N, Debatin K-M. Novel approaches to apoptosis-inducing therapies. Advances in Experimental Medicine and Biology. 2016;930:173–204.
  • Westhoff M-A, Marschall N, Grunert M, et al. Cell death-based treatment of childhood cancer. Cell Death & Disease. 2018;9:116.
  • Mahvi DA, Liu R, Grinstaff MW, et al. Local cancer recurrence: the realities, challenges, and opportunities for new therapies. CA: A Cancer Journal for Clinicians. 2018;68: 488–505.
  • Persano L, Rampazzo E, Della Puppa A, et al. The three-layer concentric model of glioblastoma: cancer stem cells, microenvironmental regulation, and therapeutic implications. TheScientificWorldJournal. 2011;11:1829–1841.
  • Grunert M, Kassubek R, Danz B, et al. Radiation and brain tumors: an overview. Critical Reviews in Oncogenesis. 2018;23:119–138.
  • Schosserer M, Grillari J, Breitenbach M. The dual role of cellular senescence in developing tumors and their response to cancer therapy. Frontiers in Oncology. 2017;7:278.
  • Little JB. Cellular, molecular, and carcinogenic effects of radiation. Hematology/oncology Clinics of North America. 1993;7:337–352.
  • Ferguson LR, Pearson AE. The clinical use of mutagenic anticancer drugs. Mutation Research. 1996;355:1–12.
  • Kattner P, Strobel H, Khoshnevis N, et al. Compare and contrast: pediatric cancer versus adult malignancies. Cancer Metastasis Reviews. 2019;38:673–682.
  • Thorburn AL. Paul ehrlich: pioneer of chemotherapy and cure by arsenic (1854-1915). The British Journal of Venereal Diseases. 1983;59:404–405.
  • Chabner BA, Roberts TG. Timeline: chemotherapy and the war on cancer. Nature Reviews. Cancer. 2005;5:65–72.
  • Mansoori B, Mohammadi A, Davudian S, et al The different mechanisms of cancer drug resistance: a brief review. Advanced Pharmaceutical Bulletin. 2017;7:339–348.
  • Strobel H, Baisch T, Fitzel R, et al Temozolomide and Other Alkylating Agents in Glioblastoma Therapy. Biomedicines. 2019;7. DOI:10.3390/biomedicines7030069
  • Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nature Medicine. 1996;2:561–566.
  • Vaerman JL, Lewalle P, Martiat P. Antisense inhibition of P210 bcr-abl in chronic myeloid leukemia. Stem Cells (Dayton, Ohio). 1993;89–95. DOI:10.1002/stem.5530110921
  • McDonald ES, Clark AS, Tchou J, et al Clinical diagnosis and management of breast cancer. Journal of Nuclear Medicine: Official Publication, Society of Nuclear Medicine 2016 9S–16S. DOI: 10.2967/jnumed.115.157834
  • Lei S, Zhang X, Li J, et al. Current progress in messenger RNA-based gene therapy. Journal of Biomedical Nanotechnology. 2020;16:1018–1044.
  • Crooke ST, Witztum JL, Bennett CF, et al. RNA-Targeted Therapeutics. Cell Metabolism. 2018;27:714–739.
  • Chhipa AS, Patel S. Targeting pyruvate kinase muscle isoform 2 (PKM2) in cancer: what do we know so far? Life Sciences. 2021;280:119694.
  • Sahin U, Oehm P, Derhovanessian E, et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature. 2020;585:107–112.
  • Esfahani K, Roudaia L, Buhlaiga N, et al. A review of cancer immunotherapy: from the past, to the present, to the future. Current Oncology (Toronto, Ont.). 2020;27:S87–S97.
  • Pantin J, Battiwalla M. Upsetting the apple CAR-T (chimeric antigen receptor T-cell therapy) - sustainability mandates USA innovation. British Journal of Haematology. 2020;190:851–853.
  • Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. International Immunopharmacology. 2018;62:29–39.
  • van den Bulk J, Verdegaal EM, de Miranda NF. Cancer immunotherapy: broadening the scope of targetable tumours. Open Biol. 2018;8(6):180037. DOI:10.1098/rsob.180037.
  • Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their Inhibition. American Journal of Clinical Oncology. 2016;39:98–106.
  • Abbott M, Ustoyev Y. Cancer and the immune system: the history and background of immunotherapy. Seminars in Oncology Nursing. 2019;35:150923.
  • Gong J, Chehrazi-Raffle A, Reddi S, et al. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer. 2018;6.
  • Cameron D, Piccart-Gebhart MJ, Gelber RD, et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the herceptin adjuvant (HERA) trial. Lancet (London, England). 2017;389:1195–1205.
  • Gerratana L, Bonotto M, Bozza C, et al. Pertuzumab and breast cancer: another piece in the anti-HER2 puzzle. Expert Opinion on Biological Therapy. 2017;17:365–374.
  • Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer Journal. 2021;11:69.
  • Macedo N, Miller DM, Haq R, et al. Clinical landscape of oncolytic virus research in 2020. J Immunother Cancer. 2020;8(2):e001486.
  • Faghfuri E, Pourfarzi F, Faghfouri AH, et al. Recent developments of RNA-based vaccines in cancer immunotherapy. Expert Opinion on Biological Therapy. 2021;21:201–218.
  • Van Nuffel AMT, Wilgenhof S, Thielemans K, et al. Overcoming HLA restriction in clinical trials: immune monitoring of mRNA-loaded DC therapy. Oncoimmunology. 2012;1:1392–1394.
  • Stanković T, Dinić J, Podolski-Renić A, et al. Dual Inhibitors as a new challenge for cancer multidrug resistance treatment. Curr Med Chem. 2019;26:6074–6106.
  • Fischer T, Najjar A, Totzke F, et al. Discovery of novel dual inhibitors of receptor tyrosine kinases egfr and pdgfr-β related to anticancer drug resistance. J Enzyme Inhib Med Chem. 2018;33:1–8.
  • Quesnelle KM, Grandis JR. Dual Kinase Inhibition of EGFR and HER2 overcomes resistance to cetuximab in a novel in vivo model of acquired cetuximab resistance. Clin Cancer Res. 2011;17:5935–5944.
  • Smith LK, Sheppard KE, McArthur GA. Is resistance to targeted therapy in cancer inevitable? Cancer Cell. 2021;39:1047–1049.
  • Loeb LA, Springgate CF, Battula N. Errors in DNA replication as a basis of malignant changes. Cancer Research. 1974;34:2311–2321.
  • Vendramin R, Litchfield K, Swanton C. Cancer evolution: Darwin and beyond. The EMBO Journal. 2021;40:e108389.
  • Lovely CM, Shaw AT. Molecular pathways: resistance to kinase inhibitors and implications for therapeutic strategies. Clin Cancer Res. 2014;20:2249–2256.
  • Kuosmanen T, Cairns J, Noble R, et al. Drug-induced resistance evolution necessitates less aggressive treatment. PLoS Computational Biology. 2021;17:e1009418.
  • Jiapaer S, Furuta T, Tanaka S, et al. potential strategies overcoming the temozolomide resistance for glioblastoma. Neurologia medico-chirurgica. 2018;58:405–421.
  • Haar CP, Hebbar P, Wallace GC, et al. Drug resistance in glioblastoma: a mini review. Neurochemical Research. 2012;37:1192–1200.
  • Martínez A, Sesé M, Losa JH, et al. Phosphorylation of eIF4E confers resistance to cellular stress and dna-damaging agents through an interaction with 4E-T: a rationale for novel therapeutic approaches. PloS one. 2015;10:e0123352.
  • Fulda S, Gorman AM, Hori O, et al. Cellular stress responses: cell survival and cell death. International Journal of Cell Biology. 2010;2010:214074.
  • Batlle E, Massagué J. Transforming growth factor-β signaling in immunity and cancer. Immunity. 2019;50:924–940.
  • Vaziri C, Rogozin IB, Gu Q, et al. Unravelling roles of error-prone DNA polymerases in shaping cancer genomes. Oncogene. 2021;40:6549–6565.
  • Aderemi AO, Novais SC, Lemos MFL, et al. Oxidative stress responses and cellular energy allocation changes in microalgae following exposure to widely used human antibiotics. Aquatic Toxicology (Amsterdam, Netherlands). 2018;203:130–139.
  • Loeb LA. Mutator phenotype in cancer: origin and consequences. Semin Cancer Biol. 2010;20:279–280.
  • Lee MJ, Ye AS, Gardino AK, et al. Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks. Cell. 2012;149:780–794.
  • Park C, Qian W, Zhang J. Genomic evidence for elevated mutation rates in highly expressed genes. EMBO Rep. 2012;13:1123–1129.
  • Makova KD, Hardison RC. The Effects of Chromatin Organization on Variation in Mutation Rates in the Genome. Nat Rev Genet. 2015;16:213–223.
  • Akdemir KC, Le VT, Kim JM, et al. Somatic mutation distributions in cancer genomes vary with three-dimensional chromatin structure. Nat Genet. 2020;52:1178–1188.
  • Monroe JG, Srikant T, Carbonell-Bejerano P, et al. Mutation Bias Reflects Natural Selection in Arabidopsis Thaliana. Nature. 2022;602:101–105.
  • Alexandrov LB, Kim J, Haradhvala NJ, et al. The repertoire of mutational signatures in human cancer. Nature. 2020;578:94–101.
  • Boffetta P, Kaldor JM. Secondary malignancies following cancer chemotherapy. Acta Oncol. 1994;33:591–598.
  • Cheung-Ong K, Giaever G, Nislow C. DNA-Damaging agents in cancer chemotherapy: serendipity and chemical biology. Chem Biol. 2013;20:648–659.
  • Westhoff M, Brühl O, Debatin K. Cancer therapy: know your enemy? Mol Cell Pediatr. 2014;1:10.
  • Shapiro L, McAdams HH, Losick J. Generating and exploiting polarity in bacteria. Science. 2002;298:1942–1946.
  • Fitzgerald DM, Hastings PJ, Rosenberg SM. Stress-induced mutagenesis: implications in cancer and drug resistance. Annu Rev Cancer Biol. 2017;1:119–140.
  • Luria SE, Delbrück M. Mutations of bacteria from virus sensitivity to virus resistance. Genetics. 1943;28:491–511.
  • Melnikov SV, Stevens DL, Fu X, et al. Exploiting evolutionary trade-offs for posttreatment management of drug-resistant populations. Proc Natl Acad Sci USA. 2020;117:17924–17931.
  • Nichol D, Jeavons P, Fletcher AG, et al. Steering evolution with sequential therapy to prevent the emergence of bacterial antibiotic resistance. PLoS Comput Biol. 2015;11:e1004493.
  • Udekwu KI, Weiss H. Pharmacodynamic considerations of collateral sensitivity in design of antibiotic treatment regimen. Drug Des Devel Ther. 2018;12:2249–2257.
  • Imamovic L, Sommer MOA. Use of collateral sensitivity networks to design drug cycling protocols that avoid resistance development. Sci Transl Med. 2013;5:204ra132.
  • Palmer AC, Chidley C, Sorger PK. A curative combination cancer therapy achieves high fractional cell killing through low cross-resistance and drug additivity. Elife. 2019;8:e50036.
  • Roemhild R, Gokhale CS, Dirksen P, et al. Cellular hysteresis as a principle to maximize the efficacy of antibiotic therapy. Proc Natl Acad Sci USA. 2018;115:9767–9772.
  • Veening J-W, Smits WK, Kuipers OP. Bistability, epigenetics, and bet-hedging in bacteria. Annu Rev Microbiol. 2008;62:193–210.
  • Herrmann MD, Lennerz JK, Bullinger L, et al. Transitory dasatinib-resistant states in KIT(mut) t(8;21) acute myeloid leukemia cells correlate with altered KIT expression. Exp Hematol. 2014; 42: 90–100.
  • Batra A, Roemhild R, Rousseau E, et al. High potency of sequential therapy with only β-lactam antibiotics. Elife. 2021;10. DOI:10.7554/eLife.68876
  • Barbosa C, Römhild R, Rosenstiel P, et al. Evolutionary stability of collateral sensitivity to antibiotics in the model pathogen Pseudomonas aeruginosa. Elife. 2019;8.
  • Bosanquet AG, Bell PB. Enhanced ex vivo drug sensitivity testing of chronic lymphocytic leukaemia using refined DiSC assay methodology. Leuk Res. 1996;20:143–153.
  • Scarborough JA, McClure E, Anderson P, et al. Identifying states of collateral sensitivity during the evolution of therapeutic resistance in ewing’s sarcoma. iScience. 2020;23:101293.
  • Doherty B, Lawlor D, Gillet J-P, et al. Collateral sensitivity to cisplatin in KB-8-5-11 drug-resistant cancer cells. Anticancer Res. 2014;34:503–507.
  • Gatenby RA, Silva AS, Gillies RJ, et al. Adaptive therapy. Cancer Res. 2009;69:4894–4903.
  • Vrankar M, Stanic K. Long-term survival of locally advanced stage III non-small cell lung cancer patients treated with chemoradiotherapy and perspectives for the treatment with immunotherapy. Radiol Oncol. 2018;52:281–288.
  • Sytov A, Brenin C, Millard T, et al. Long-Term non-progression in metastatic breast cancer beyond 5 years: case series and review. Curr Breast Cancer Rep. 2021;13:208–215.
  • Le Tourneau C, Delord J-P, Gonçalves A, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–1334.
  • Gambardella V, Tarazona N, Cejalvo JM, et al. Personalized medicine: recent progress in cancer therapy. Cancers (Basel). 2020;12(4):1009.
  • Hirata E, Girotti MR, Viros A, et al. Intravital imaging reveals how BRAF inhibition generates drug-tolerant microenvironments with high integrin β1/FAK signaling. Cancer Cell. 2015;27:574–588.
  • Fiedler EC, Hemann MT. Aiding and abetting: how the tumor microenvironment protects cancer from chemotherapy. Annu Rev Cancer Biol. 2019;3:409–428.
  • Geller LT, Barzily-Rokni M, Danino T, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156–1160.
  • Hong DS, Vence L, Falchook G, et al. BRAF(V600) inhibitor GSK2118436 targeted inhibition of mutant BRAF in cancer patients does not impair overall immune competency. Clin Cancer Res. 2012;18:2326–2335.
  • Gambacorti-Passerini C, Antolini L, Mahon F-X, et al. Multicenter independent assessment of outcomes in chronic myeloid leukemia patients treated with imatinib. J Natl Cancer Inst. 2011;103:553–561.
  • Luebker SA, Koepsell SA. Diverse mechanisms of braf inhibitor resistance in melanoma identified in clinical and preclinical studies. Front Oncol. 2019;9:268.
  • Hochhaus A, Kreil S, Corbin AS, et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia. 2002;16:2190–2196.
  • Corbacioglu S, Steinbach D, Lode HN, et al. The RIST design: a molecularly targeted multimodal approach for the treatment of patients with relapsed and refractory neuroblastoma. JCO. 2013; 31:10017.
  • Indolfi P, Corbacioglu S, Perrotta S, et al. Targeted molecular therapy (modified RIST regimen) in relapsed high risk stage IV neuroblastoma: two cases report. 2018.
  • Nonnenmacher L, Westhoff M-A, Fulda S, et al. RIST: a potent new combination therapy for glioblastoma. Int J Cancer. 2015;136:E173–187.
  • Westhoff M-A, Faham N, Marx D, et al. Sequential dosing in chemosensitization: targeting the PI3K/Akt/mTOR pathway in neuroblastoma. PLoS One. 2013;8:e83128.
  • Hasslacher S, Schneele L, Stroh S, et al. Inhibition of PI3K signalling increases the efficiency of radiotherapy in glioblastoma cells. Int J Oncol. 2018;53:1881–1896.
  • Mettang M, Meyer-Pannwitt V, Karpel-Massler G, et al. Blocking distinct interactions between glioblastoma cells and their tissue microenvironment: a novel multi-targeted therapeutic approach. Sci Rep. 2018;8:5527.
  • Zeyen T, Potthoff A-L, Nemeth R, et al. Phase I/II trial of meclofenamate in progressive MGMT-methylated glioblastoma under temozolomide second-line therapy-the mecmeth/NOA-24 trial. Trials. 2022;23:57.
  • Halatsch M-E, Dwucet A, Schmidt CJ, et al. In vitro and clinical compassionate use experiences with the drug-repurposing approach cusp9v3 in glioblastoma. Pharmaceuticals. 2021;14(12):1241.
  • Kragh H. From disulfiram to antabuse: the invention of a drug. Bull Hist Chem. 2008;33:82–88.
  • Sanny CG, Weiner H. Interaction of disulfiram with horse liver aldehyde dehydrogenase, in alcohol and aldehyde metabolizing systems. Academic Press. 1977:167–174.
  • Langeland BT, McKinley-McKee JS. The effects of disulfiram and related compounds on equine hepatic alcohol dehydrogenase. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol. 1997;117:55–61.
  • Schreck R, Meier B, Männel DN, et al. Dithiocarbamates as potent inhibitors of nuclear factor kappa B activation in intact cells. J Exp Med. 1992;175:1181–1194.
  • Yang Q, Yao Y, Li K, et al. An updated review of disulfiram: molecular targets and strategies for cancer treatment. Curr Pharm Des. 2019;25:3248–3256.
  • Sirota M, Dudley JT, Kim J, et al. Discovery and preclinical validation of drug indications using compendia of public gene expression data. Sci Transl Med. 2011;3:96ra77.
  • Siegelin MD, Schneider E, Westhoff M-A, et al. Current state and future perspective of drug repurposing in malignant glioma. Semin Cancer Biol. 2021;68:92–104.
  • Farrier D. Humanity’s influence on living organisms – both intentional and unintentional – is causing them to evolve in new and unusual ways. But how far could human-driven adaptation go? [Internet]. 2022. [cited 2022 Feb 2]. Available from: https://www.bbc.com/future/article/20220125-how-humans-are-changing-evolution
  • Ferlay J, Ervik M, Colomet M, et al. Global cancer observatory: cancer today. Lyon: International Agency for Research on Cancer [Internet]. [cited 2022 Feb 19]. Available from: https://gco.iarc.fr/today
  • Ghosal G, Chen J. DNA damage tolerance: a double-edged sword guarding the genome. Transl Cancer Res. 2013;2:107–129.
  • Hientz K, Mohr A, Bhakta-Guha D, et al. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget. 2017;8:8921–8946.
  • Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
  • Gainor JF, Dardaei L, Yoda S, et al. Molecular mechanisms of resistance to first- and second-generation alk inhibitors in ALK-rearranged lung cancer. Cancer Discov. 2016;6:1118–1133.
  • Dong X, Fernandez-Salas E, Li E, et al. Elucidation of resistance mechanisms to second-generation alk inhibitors alectinib and ceritinib in non-small cell lung cancer cells. Neoplasia. 2016;18:162–171.
  • Carlson RD, Flickinger JC, Snook AE. Talkin’ toxins: from coley’s to modern cancer immunotherapy. Toxins (Basel). 2020;13:12.
  • Taefehshokr S, Parhizkar A, Hayati S, et al. Cancer immunotherapy: challenges and limitations. Pathol Res Pract. 2022;229:153723.
  • Gröbner SN, Worst BC, Weischenfeldt J, et al. The landscape of genomic alterations across childhood cancers. Nature. 2018;555:321–327.
  • Blaeschke F, Paul MC, Schuhmann MU, et al. Low mutational load in pediatric medulloblastoma still translates into neoantigens as targets for specific T-cell immunotherapy. Cytotherapy. 2019;21:973–986.
  • Zhao J, Chen AX, Gartrell RD, et al. Author Correction: immune and Genomic Correlates of Response to Anti-PD-1 Immunotherapy in Glioblastoma. Nat Med. 2019;25:1022.
  • Van Gool SW, Makalowski J, Bitar M, et al. Synergy between TMZ and individualized multimodal immunotherapy to improve overall survival of idh1 wild-type MGMT promoter-unmethylated gbm patients. Genes Immun. 2022. DOI:10.1038/s41435-022-00162-y

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.