Ecological and evolutionary dynamics to design and improve ovarian cancer treatment

Grace Y. Q. Han , Monica Alexander , Julia Gattozzi , Marilyn Day , Elayna Kirsch , Narges Tafreshi , Raafat Chalar , Soraya Rahni , Gabrielle Gossner , William Burke , Mehdi Damaghi

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (9) : e70012

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (9) : e70012 DOI: 10.1002/ctm2.70012
REVIEW

Ecological and evolutionary dynamics to design and improve ovarian cancer treatment

Author information +
History +
PDF

Abstract

•Tumours are ecosystems in which cancer and non-cancer cells interact and evolve in complex and dynamic ways.

•Conventional therapies for ovarian cancer inevitably lead to the development of resistance because they fail to consider tumours’ heterogeneity and cellular plasticity.

•Eco-evolutionarily designed therapies should consider cancer cell plasticity and patient-specific characteristics to improve clinical outcome and prevent relapse.

Keywords

ovarian cancer / phenotypic plasticity / tumour ecology / tumour evolution / tumour microenvironment

Cite this article

Download citation ▾
Grace Y. Q. Han, Monica Alexander, Julia Gattozzi, Marilyn Day, Elayna Kirsch, Narges Tafreshi, Raafat Chalar, Soraya Rahni, Gabrielle Gossner, William Burke, Mehdi Damaghi. Ecological and evolutionary dynamics to design and improve ovarian cancer treatment. Clinical and Translational Medicine, 2024, 14(9): e70012 DOI:10.1002/ctm2.70012

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

SiegelRL, MillerKD, WagleNS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023; 73: 17-48

[2]

SaaniI, RajN, SoodR, et al. Clinical challenges in the management of malignant ovarian germ cell tumours. Int J Environ Res Public Health. 2023; 20: 6089.

[3]

LiH, LiuY, WangY, et al. Hormone therapy for ovarian cancer: emphasis on mechanisms and applications (review). Oncol Rep. 2021; 46: 223.

[4]

Tomás-PérezS, OtoJ, Aghababyan C, et al. Increased levels of NETosis biomarkers in high-grade serous ovarian cancer patients’ biofluids: potential role in disease diagnosis and management. Front Immunol. 2023; 14:1111344

[5]

GhoseA, BolinaA, MahajanI, et al. Hereditary ovarian cancer: towards a cost-effective prevention strategy. Int J Environ Res Public Health. 2022; 19: 12057.

[6]

Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Ovarian Cancer Including Fallopian Tube Cancer and Primary Peritoneal Cancer V.3.2024. National Comprehensive Cancer Network, Inc. 2024. Accessed July 31, 2024. www.NCCN.org.

[7]

ChristieEL, Bowtell DDL. Acquired chemotherapy resistance in ovarian cancer. Ann Oncol. 2017; 28: viii13-viii15.

[8]

LiC-J, LinL-T, TsaiH-W, et al. The molecular regulation in the pathophysiology in ovarian aging. Aging Dis. 2021; 12: 934–949

[9]

LipinskiKA, BarberLJ, DaviesMN, et al. Cancer evolution and the limits of predictability in precision cancer medicine. Trends Cancer Res. 2016; 2: 49-63

[10]

MatulonisUA, SoodAK, FallowfieldL, et al. Ovarian cancer. Nat Rev Dis Primers. 2016; 2:16061

[11]

EngbersenMP, Van Driel W, LambregtsD, LahayeM. The role of CT, PET-CT, and MRI in ovarian cancer. Br J Radiol. 2021; 94:20210117

[12]

ZhangM, ChengS, JinY, et al. Roles of CA125 in diagnosis, prediction, and oncogenesis of ovarian cancer. Biochim Biophys Acta Rev Cancer. 2021; 1875:188503

[13]

GhoseA, McCannL, MakkerS, et al. Diagnostic biomarkers in ovarian cancer: advances beyond CA125 and HE4. Ther Adv Med Oncol. 2024; 16:17588359241233225

[14]

MaloneySM, HooverCA, Morejon-LassoLV, ProsperiJR. Mechanisms of taxane resistance. Cancers. 2020; 12: 3323.

[15]

SmithER, WangJ-Q, YangD-H, Xu X-X. Paclitaxel resistance related to nuclear envelope structural sturdiness. Drug Resist Updat. 2022; 65:100881

[16]

Gynecologic Oncology Group, Markman M, BlessingJ, et al. Phase II trial of weekly paclitaxel (80 mg/m2) in platinum and paclitaxel-resistant ovarian and primary peritoneal cancers: a Gynecologic Oncology Group study. Gynecol Oncol. 2006; 101: 436–440

[17]

ZhouJ, KangY, ChenL, et al. The drug-resistance mechanisms of five platinum-based antitumor agents. Front Pharmacol. 2020; 11: 343

[18]

TchounwouPB, DasariS, NoubissiFK, et al. Advances in our understanding of the molecular mechanisms of action of cisplatin in cancer therapy. J Exp Pharmacol. 2021; 13: 303–328

[19]

RottenbergS, DislerC, PeregoP. The rediscovery of platinum-based cancer therapy. Nat Rev Cancer. 2021; 21: 37–50

[20]

AronsonSL, Lopez-Yurda M, KooleSN, et al. Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy in patients with advanced ovarian cancer (OVHIPEC-1): final survival analysis of a randomised, controlled, phase 3 trial. Lancet Oncol. 2023; 24: 1109–1118

[21]

SpiliotisJ, HalkiaE, LianosE, et al. Cytoreductive surgery and HIPEC in recurrent epithelial ovarian cancer: a prospective randomized phase III study. Ann Surg Oncol. 2015; 22: 1570–1575

[22]

ShawP, Dwivedi SKD, BhattacharyaR, et al. VEGF signaling: role in angiogenesis and beyond. Biochim Biophys Acta Rev Cancer. 2024; 1879:189079

[23]

HaibeY, Kreidieh M, El HajjH, et al. Resistance mechanisms to anti-angiogenic therapies in cancer. Front Oncol. 2020; 10: 221

[24]

HorikawaN, AbikoK, MatsumuraN, et al. Anti-VEGF therapy resistance in ovarian cancer is caused by GM-CSF-induced myeloid-derived suppressor cell recruitment. Br J Cancer. 2020; 122: 778–788

[25]

WangL, LiangC, LiF, et al. PARP1 in carcinomas and PARP1 inhibitors as antineoplastic drugs. Int J Mol Sci. 2017; 18: 2111.

[26]

CaronM-C, SharmaAK, O’SullivanJ, et al. Poly(ADP-ribose) polymerase-1 antagonizes DNA resection at double-strand breaks. Nat Commun. 2019; 10: 2954

[27]

YoonW-H, DeFazio A, KashermanL. Immune checkpoint inhibitors in ovarian cancer: where do we go from here? Cancer Drug Resist. 2023; 6: 358–377

[28]

WuY, XuS, ChengS, et al. Clinical application of PARP inhibitors in ovarian cancer: from molecular mechanisms to the current status. J Ovarian Res. 2023; 16: 6

[29]

PiombinoC, Cortesi L. Insights into the possible molecular mechanisms of resistance to PARP inhibitors. Cancers. 2022; 14: 2804.

[30]

LordCJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017; 355: 1152–1158

[31]

DiasMP, MoserSC, GanesanS, Jonkers J. Understanding and overcoming resistance to PARP inhibitors in cancer therapy. Nat Rev Clin Oncol. 2021; 18: 773–791

[32]

MooreK, Colombo N, ScambiaG, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2018; 379: 2495–2505

[33]

MirzaMR, MonkBJ, HerrstedtJ, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 2016; 375: 2154–2164

[34]

González-MartínA, PothuriB, Vergote I, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2019; 381: 2391–2402

[35]

WangY, DuvalAJ, AdliM, Matei D. Biology-driven therapy advances in high-grade serous ovarian cancer. J Clin Invest. 2024; 134: e174013.

[36]

KristeleitRS, MooreKN. Life after SOLO-2: is olaparib really inducing platinum resistance in BRCA-mutated (BRCAm), PARP inhibitor (PARPi)-resistant, recurrent ovarian cancer? Ann Oncol. 2022; 33: 989–991

[37]

KimD-S, Camacho CV, KrausWL. Alternate therapeutic pathways for PARP inhibitors and potential mechanisms of resistance. Exp Mol Med. 2021; 53: 42–51

[38]

LuoH, LiS, ZhaoM, et al. Prognostic value of progesterone receptor expression in ovarian cancer: a meta-analysis. Oncotarget. 2017; 8: 36845–36856

[39]

ShenZ, LuoH, LiS, et al. Correlation between estrogen receptor expression and prognosis in epithelial ovarian cancer: a meta-analysis. Oncotarget. 2017; 8: 62400–62413

[40]

BorellaF, FucinaS, MangheriniL, et al. Hormone receptors and epithelial ovarian cancer: recent advances in biology and treatment options. Biomedicines. 2023; 11: 2157.

[41]

YangY, YangY, YangJ, et al. Tumor microenvironment in ovarian cancer: function and therapeutic strategy. Front Cell Dev Biol. 2020; 8: 758

[42]

GiraldoNA, Sanchez-Salas R, PeskeJD, et al. The clinical role of the TME in solid cancer. Br J Cancer. 2019; 120: 45–53

[43]

HartnettEG, KnightJ, RadolecM, et al. Immunotherapy advances for epithelial ovarian cancer. Cancers. 2020; 12: 3733.

[44]

GhisoniE, Imbimbo M, ZimmermannS, ValabregaG. Ovarian cancer immunotherapy: turning up the heat. Int J Mol Sci. 2019; 20: 2927.

[45]

WagnerJ, Wickman E, DeRenzoC, GottschalkS. CAR T cell therapy for solid tumors: bright future or dark reality? Mol Ther. 2020; 28: 2320–2339

[46]

YangS, YinX, YueY, WangS. Application of adoptive immunotherapy in ovarian cancer. Onco Targets Ther. 2019; 12: 7975–7991

[47]

RosenbergSA, YangJC, SherryRM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011; 17: 4550–4557

[48]

KeamSJ. Lifileucel: first approval. Mol Diagn Ther. 2024; 28: 339–344

[49]

FanaleD, DiminoA, PedoneE, et al. Prognostic and predictive role of tumor-infiltrating lymphocytes (TILs) in ovarian cancer. Cancers. 2022; 14: 4344.

[50]

Ovarian Tumor Tissue Analysis (OTTA) Consortium, GoodeEL, BlockMS, et al. Dose–response association of CD8+ tumor-infiltrating lymphocytes and survival time in high-grade serous ovarian cancer. JAMA Oncol. 2017; 3: e173290

[51]

JindalV, AroraE, GuptaS, et al. Prospects of chimeric antigen receptor T cell therapy in ovarian cancer. Med Oncol. 2018; 35: 70

[52]

ShahNN, FryTJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol. 2019; 16: 372–385

[53]

KatzSC, PointGR, CunettaM, et al. Regional CAR-T cell infusions for peritoneal carcinomatosis are superior to systemic delivery. Cancer Gene Ther. 2016; 23: 142–148

[54]

Cutri-FrenchC, Nasioudis D, GeorgeE, TanyiJL. CAR-T cell therapy in ovarian cancer: where are we now? Diagnostics (Basel). 2024; 14: 819.

[55]

BauluE, GardetC, ChuvinN, Depil S. TCR-engineered T cell therapy in solid tumors: state of the art and perspectives. Sci Adv. 2023; 9: eadf3700

[56]

WuJWY, DandS, DoigL, et al. T-cell receptor therapy in the treatment of ovarian cancer: a mini review. Front Immunol. 2021; 12:672502

[57]

DemircanNC, Boussios S, TasciT, ÖztürkMA. Current and future immunotherapy approaches in ovarian cancer. Ann Transl Med. 2020; 8: 1714

[58]

GubinMM, Artyomov MN, MardisER, SchreiberRD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015; 125: 3413–3421

[59]

GoodmanAM, KatoS, BazhenovaL, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017; 16: 2598–2608

[60]

YenY-T, ChienM, WuP-Y, et al. Protein phosphatase 2A inactivation induces microsatellite instability, neoantigen production and immune response. Nat Commun. 2021; 12: 7297

[61]

HinchcliffE, PatelA, FellmanB, et al. Loss-of-function mutations in PPP2R1A correlate with exceptional survival in ovarian clear cell carcinomas treated with immune checkpoint inhibitors (099). Gynecol Oncol. 2022; 166: S66

[62]

ClinicalTrials.Gov. Accessed July 31, 2024. https://www.clinicaltrials.gov/study/NCT06065462

[63]

FranzeseO, Graziani G. Role of PARP inhibitors in cancer immunotherapy: potential friends to immune activating molecules and foes to immune checkpoints. Cancers. 2022; 14: 5633.

[64]

MarthC, Park-Simon T-W, AghajanianC, et al. #1120 Durvalumab with paclitaxel/carboplatin + bevacizumab then maintenance durvalumab, bevacizumab + olaparib in patients with newly diagnosed advanced ovarian cancer without a tumour BRCA1/2 mutation: results from the DUO-O/ENGOT-Ov46/AGO-OVAR 23/GOG-3025 trial. Late Breaking Abstracts. BMJ Publishing Group Ltd. 2023.

[65]

LiuZ, ZhouZ, DangQ, et al. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Theranostics. 2022; 12: 6273–6290

[66]

ChengJ, ZhaoL, ZhangY, et al. Understanding the mechanisms of resistance to CAR T-cell therapy in malignancies. Front Oncol. 2019; 9: 1237

[67]

PengW, ChenJQ, LiuC, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov. 2016; 6: 202–216

[68]

GroellyFJ, FawkesM, DaggRA, et al. Targeting DNA damage response pathways in cancer. Nat Rev Cancer. 2023; 23: 78–94

[69]

LorussoD, Mouret-Reynier M-A, HarterP, et al. Updated progression-free survival and final overall survival with maintenance olaparib plus bevacizumab according to clinical risk in patients with newly diagnosed advanced ovarian cancer in the phase III PAOLA-1/ENGOT-ov25 trial. Int J Gynecol Cancer. 2024; 34: 550–558

[70]

ClinicalTrials.Gov. Accessed July 31, 2024. https://www.clinicaltrials.gov/study/NCT03740165

[71]

ClinicalTrials.Gov. Accessed July 31, 2024. https://www.clinicaltrials.gov/study/NCT03737643

[72]

ClinicalTrials.Gov. Accessed July 31, 2024. https://www.clinicaltrials.gov/study/NCT03587311

[73]

ClinicalTrials.Gov. Accessed July 31, 2024. https://www.clinicaltrials.gov/study/NCT03353831

[74]

HuangT-T, Lampert EJ, CootsC, LeeJ-M. Targeting the PI3K pathway and DNA damage response as a therapeutic strategy in ovarian cancer. Cancer Treat Rev. 2020; 86:102021

[75]

KimH, XuH, GeorgeE, et al. Combining PARP with ATR inhibition overcomes PARP inhibitor and platinum resistance in ovarian cancer models. Nat Commun. 2020; 11: 3726

[76]

BoussiosS, Karathanasi A, CookeD, et al. PARP inhibitors in ovarian cancer: the route to “Ithaca.” Diagnostics (Basel). 2019; 9: 55.

[77]

ZielliT, Labidi-Galy I, Del GrandeM, et al. The clinical challenges of homologous recombination proficiency in ovarian cancer: from intrinsic resistance to new treatment opportunities. Cancer Drug Resist. 2023; 6: 499–516

[78]

XuL, SunH, LemoineNR, et al. Oncolytic vaccinia virus and cancer immunotherapy. Front Immunol. 2023; 14:1324744

[79]

LiM, ZhangM, YeQ, et al. Preclinical and clinical trials of oncolytic vaccinia virus in cancer immunotherapy: a comprehensive review. Cancer Biol Med. 2023; 20: 646–661

[80]

ClinicalTrials.Gov. Accessed July 31, 2024. https://www.clinicaltrials.gov/study/NCT05281471

[81]

Genelux Corporation Receives FDA Fast Track Designation for Olvi-Vec in Platinum Resistant/Refractory Ovarian Cancer. Genelux Corporation. Accessed July 31, 2024. https://investors.genelux.com/news-releases/news-release-details/genelux-corporation-receives-fda-fast-track-designation-olvi-vec

[82]

BaxHJ, Chauhan J, StavrakaC, et al. Folate receptor alpha in ovarian cancer tissue and patient serum is associated with disease burden and treatment outcomes. Br J Cancer. 2023; 128: 342–353

[83]

HeoY-A. Mirvetuximab soravtansine: first approval. Drugs. 2023; 83: 265–273

[84]

PonteJF, AbO, LanieriL, et al. Mirvetuximab Soravtansine (IMGN853), a folate receptor alpha-targeting antibody-drug conjugate, potentiates the activity of standard of care therapeutics in ovarian cancer models. Neoplasia. 2016; 18: 775–784

[85]

GilbertL, OakninA, MatulonisUA, et al. Safety and efficacy of mirvetuximab soravtansine, a folate receptor alpha (FRα)-targeting antibody-drug conjugate (ADC), in combination with bevacizumab in patients with platinum-resistant ovarian cancer. Gynecol Oncol. 2023; 170: 241–247

[86]

BaileyC, BlackJRM, ReadingJL, et al. Tracking cancer evolution through the disease course. Cancer Discov. 2021; 11: 916–932

[87]

MartinsFC, Couturier D-L, de SantiagoI, et al. Clonal somatic copy number altered driver events inform drug sensitivity in high-grade serous ovarian cancer. Nat Commun. 2022; 13: 6360

[88]

FrançaGS, BaronM, KingBR, et al. Cellular adaptation to cancer therapy along a resistance continuum. Nature. 2024; 631: 876–883

[89]

RaeC, AmatoF, BraconiC. Patient-derived organoids as a model for cancer drug discovery. Int J Mol Sci. 2021; 22: 3483.

[90]

LuZ, NieB, ZhaiW, Hu Z. Delineating the longitudinal tumor evolution using organoid models. J Genet Genomics. 2021; 48: 560–570

[91]

DattaP, DeyM, AtaieZ, et al. 3D bioprinting for reconstituting the cancer microenvironment. NPJ Precis Oncol. 2020; 4: 18

[92]

MaenhoudtN, Vankelecom H. Protocol for establishing organoids from human ovarian cancer biopsies. STAR Protoc. 2021; 2:100429

[93]

VenezianiAC, Gonzalez-Ochoa E, AlqaisiH, et al. Heterogeneity and treatment landscape of ovarian carcinoma. Nat Rev Clin Oncol. 2023; 20: 820–842

[94]

HofL, MorethT, KochM, et al. Long-term live imaging and multiscale analysis identify heterogeneity and core principles of epithelial organoid morphogenesis. BMC Biol. 2021; 19: 37

[95]

MarineJ-C, DawsonS-J, DawsonMA. Non-genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer. 2020; 20: 743–756

[96]

Fernandez-MateosJ, Milite S, OliveiraE, et al. Epigenetic heritability of cell plasticity drives cancer drug resistance through one-to-many genotype to phenotype mapping. bioRxiv. 2023. 2023.11.15.567140.

[97]

DamaghiM, WestJ, Robertson-TessiM, et al. The harsh microenvironment in early breast cancer selects for a Warburg phenotype. Proc Natl Acad Sci U S A. 2021; 118: e2011342118.

[98]

FagottiA, Ferrandina G, FanfaniF, et al. Prospective validation of a laparoscopic predictive model for optimal cytoreduction in advanced ovarian carcinoma. Am J Obstet Gynecol. 2008; 199: 642.e1–6

[99]

SanerFA-CM, Ruggeri G, SiegenthalerF, et al. Change of Fagotti score is associated with outcome after neoadjuvant chemotherapy for ovarian cancer. Int J Gynecol Cancer. 2023; 33: 1595–1601

[100]

WeinsteinIB, JoeAK. Mechanisms of disease: oncogene addiction–a rationale for molecular targeting in cancer therapy. Nat Clin Pract Oncol. 2006; 3: 448–457

[101]

HaleR, Swearer SE. Ecological traps: current evidence and future directions. Proc Biol Sci. 2016; 283: 20152647.

[102]

SavolainenO, Lascoux M, MeriläJ. Ecological genomics of local adaptation. Nat Rev Genet. 2013; 14: 807–820

[103]

SingletonKR, Crawford L, TsuiE, et al. Melanoma therapeutic strategies that select against resistance by exploiting MYC-driven evolutionary convergence. Cell Rep. 2017; 21: 2796–2812

[104]

LinKH, RutterJC, XieA, et al. Using antagonistic pleiotropy to design a chemotherapy-induced evolutionary trap to target drug resistance in cancer. Nat Genet. 2020; 52: 408–417

[105]

Sáez-AyalaM, Montenegro MF, Sánchez-Del-Campo L, et al. Directed phenotype switching as an effective antimelanoma strategy. Cancer Cell. 2013; 24: 105–119

[106]

MoufarrijS, Dandapani M, ArthoferE, et al. Epigenetic therapy for ovarian cancer: promise and progress. Clin Epigenetics. 2019; 11: 7

[107]

KobayashiH. Recent advances in understanding the metabolic plasticity of ovarian cancer: a systematic review. Heliyon. 2022; 8: e11487

[108]

ShenY-A, HongJ, AsakaR, et al. Inhibition of the MYC-regulated glutaminase metabolic axis is an effective synthetic lethal approach for treating chemoresistant ovarian cancers. Cancer Res. 2020; 80: 4514–4526

[109]

YuanL, ShengX, ClarkLH, et al. Glutaminase inhibitor compound 968 inhibits cell proliferation and sensitizes paclitaxel in ovarian cancer. Am J Transl Res. 2016; 8: 4265–4277

[110]

WangJ-J, SiuMK-Y, JiangY-X, et al. A combination of glutaminase inhibitor 968 and PD-L1 blockade boosts the immune response against ovarian cancer. Biomolecules. 2021; 11: 1749.

[111]

ReedDR, MettsJ, PressleyM, et al. An evolutionary framework for treating pediatric sarcomas. Cancer. 2020; 126: 2577–2587

[112]

BoumahdiS, de Sauvage FJ. The great escape: tumour cell plasticity in resistance to targeted therapy. Nat Rev Drug Discov. 2020; 19: 39–56

[113]

LeighowSM, Reynolds JA, SokirniyI, et al. Programming tumor evolution with selection gene drives to proactively combat drug resistance. Nat Biotechnol. 2024. doi:10.1038/s41587-024-02271-7

[114]

GatenbyRA, SilvaAS, GilliesRJ, Frieden BR. Adaptive therapy. Cancer Res. 2009; 69: 4894–4903

[115]

ZhaoL-Y, MeiJ-X, YuG, et al. Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduct Target Ther. 2023; 8: 201

[116]

ChoiS-Y, ChoiJ-H. Ovarian cancer and the microbiome: connecting the dots for early diagnosis and therapeutic innovations—a review. Medicina. 2024; 60: 516.

[117]

ChambersLM, Esakov Rhoades EL, BhartiR, et al. Disruption of the gut microbiota confers cisplatin resistance in epithelial ovarian cancer. Cancer Res. 2022; 82: 4654–4669

[118]

HuB, SajidM, LvR, et al. A review of spatial profiling technologies for characterizing the tumor microenvironment in immuno-oncology. Front Immunol. 2022; 13:996721

[119]

SturE, Corvigno S, XuM, et al. Spatially resolved transcriptomics of high-grade serous ovarian carcinoma. iScience. 2022; 25:103923

[120]

StroblM, MartinAL, WestJ, et al. Adaptive therapy for ovarian cancer: an integrated approach to PARP inhibitor scheduling. bioRxiv. 2023. doi:10.1101/2023.03.22.533721

[121]

ClinicalTrials.Gov. Accessed August 6, 2024. https://clinicaltrials.gov/study/NCT03691376

[122]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT03017131

[123]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT05397093

[124]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT03686124

[125]

ClinicalTrials.gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT01174121

[126]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT03412877

[127]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT05194735

[128]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT05296564

[129]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT02830724

[130]

ClinicalTrials.Gov. Accessed August 6, 2024. https://www.clinicaltrials.gov/study/NCT02650986

RIGHTS & PERMISSIONS

2024 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

110

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/