Personalised, Rational, Efficacy-Driven Cancer Drug Dosing via an Artificial Intelligence SystEm (PRECISE): A Protocol for the PRECISE CURATE.AI Pilot Clinical Trial

Benjamin Kye Jyn Tan, Chong Boon Teo, Xavier Tadeo, Siyu Peng, Hazel Pei Lin Soh, Sherry De Xuan Du, Vilianty Wen Ya Luo, Aishwarya Bandla, Raghav Sundar, Dean Ho, Theodore Wonpeum Kee, Agata Blasiak, Benjamin Kye Jyn Tan, Chong Boon Teo, Xavier Tadeo, Siyu Peng, Hazel Pei Lin Soh, Sherry De Xuan Du, Vilianty Wen Ya Luo, Aishwarya Bandla, Raghav Sundar, Dean Ho, Theodore Wonpeum Kee, Agata Blasiak

Abstract

Introduction: Oncologists have traditionally administered the maximum tolerated doses of drugs in chemotherapy. However, these toxicity-guided doses may lead to suboptimal efficacy. CURATE.AI is an indication-agnostic, mechanism-independent and efficacy-driven personalised dosing platform that may offer a more optimal solution. While CURATE.AI has already been applied in a variety of clinical settings, there are no prior randomised controlled trials (RCTs) on CURATE.AI-guided chemotherapy dosing for solid tumours. Therefore, we aim to assess the technical and logistical feasibility of a future RCT for CURATE.AI-guided solid tumour chemotherapy dosing. We will also collect exploratory data on efficacy and toxicity, which will inform RCT power calculations. Methods and analysis: This is an open-label, single-arm, two-centre, prospective pilot clinical trial, recruiting adults with metastatic solid tumours and raised baseline tumour marker levels who are planned for palliative-intent, capecitabine-based chemotherapy. As CURATE.AI is a small data platform, it will guide drug dosing for each participant based only on their own tumour marker levels and drug doses as input data. The primary outcome is the proportion of participants in whom CURATE.AI is successfully applied to provide efficacy-driven personalised dosing, as judged based on predefined considerations. Secondary outcomes include the timeliness of dose recommendations, participant and physician adherence to CURATE.AI-recommended doses, and the proportion of clinically significant dose changes. We aim to initially enrol 10 participants from two hospitals in Singapore, perform an interim analysis, and consider either cohort expansion or an RCT. Recruitment began in August 2020. This pilot clinical trial will provide key data for a future RCT of CURATE.AI-guided personalised dosing for precision oncology. Ethics and dissemination: The National Healthcare Group (NHG) Domain Specific Review Board has granted ethical approval for this study (DSRB 2020/00334). We will distribute our findings at scientific conferences and publish them in peer-reviewed journals. Trial registration number: NCT04522284.

Keywords: PRECISE CURATE.AI pilot clinical trial; artificial intelligence; chemotherapy; clinical decision support system; clinical trials; oncology; personalised medicine; precision medicine.

Conflict of interest statement

DH, ABl, and TK are inventors of pending and issued patents pertaining to artificial intelligence-based drug development and personalised medicine. DH and TK are shareholders of KYAN Therapeutics, which has licensed intellectual property pertaining to AI-based drug development. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Tan, Teo, Tadeo, Peng, Soh, Du, Luo, Bandla, Sundar, Ho, Kee and Blasiak.

Figures

Figure 1
Figure 1
Internal workflow for optimising combination therapy modulation with CURATE.AI for solid tumours, including scenarios that may lead to recalibration.
Figure 2
Figure 2
Overall trial schedule for 3 weeks long cycles. BI: Baseline investigations as per standard-of-care, including collection of demographics, medical/treatment history, vital signs, and conducting complete physical examination including performance status evaluation, blood tests [haematology, serum chemistries, tumour marker(s)] and imaging as clinically required. BD: Mandatory blood draws done every 3 weeks for the measurement of tumour marker(s) between Days 17–20 of each cycle, alongside serum chemistries and haematology as per standard-of-care. BD*: Additional blood draws for measurements of tumour markers(s), done once weekly (once between Days 4–7, and another between Days 11–14) performed solely for the purposes of the trial, and are necessary for cycles 1–2 of chemotherapy. Additional blood draws in subsequent cycles will be limited to one draw, if at all. CT*: Computed tomography scans performed approximately every two or three cycles as per standard-of-care. End of study: upon completion of all chemotherapy cycles, or at 12 months, whichever is earlier for each participant.

References

    1. Peck RW. Precision medicine is not just genomics: the right dose for every patient. Annu Rev Pharmacol Toxicol. (2018) 58:105–22. 10.1146/annurev-pharmtox-010617-052446
    1. Takimoto CH. Maximum tolerated dose: clinical endpoint for a bygone era? Target Oncol. (2009) 4:143–7. 10.1007/s11523-009-0108-y
    1. Minasian L, Rosen O, Auclair D, Rahman A, Pazdur R, Schilsky RL. Optimizing dosing of oncology drugs. Clin Pharmacol Ther. (2014) 96:572–9. 10.1038/clpt.2014.153
    1. Verweij J, Casali PG, Zalcberg J, LeCesne A, Reichardt P, Blay JY, et al. . Progression-free survival in gastrointestinal stromal tumours with high-dose imatinib: randomised trial. Lancet. (2004) 364:1127–34. 10.1016/S0140-6736(04)17098-0
    1. Blanke CD, Rankin C, Demetri GD, Ryan CW, von Mehren M, Benjamin RS, et al. . Phase III randomized, intergroup trial assessing imatinib mesylate at two dose levels in patients with unresectable or metastatic gastrointestinal stromal tumors expressing the kit receptor tyrosine kinase: S0033. J Clin Oncol. (2008) 26:626–32. 10.1200/JCO.2007.13.4452
    1. Gurney H. How to calculate the dose of chemotherapy. Br J Cancer. (2002) 86:1297–302. 10.1038/sj.bjc.6600139
    1. Mathijssen RH, Sparreboom A, Verweij J. Determining the optimal dose in the development of anticancer agents. Nat Rev Clin Oncol. (2014) 11:272–81. 10.1038/nrclinonc.2014.40
    1. Gurney H. Dose calculation of anticancer drugs: a review of the current practice and introduction of an alternative. J Clin Oncol. (1996) 14:2590–611. 10.1200/JCO.1996.14.9.2590
    1. Ji Y, Jin JY, Hyman DM, Kim G, Suri A. Challenges and opportunities in dose finding in oncology and immuno-oncology. Clin Transl Sci. (2018) 11:345–51. 10.1111/cts.12540
    1. Cascorbi I. Drug interactions–principles, examples and clinical consequences. Dtsch Arztebl Int. (2012) 109:546–55. 10.3238/arztebl.2012.0546
    1. Palleria C, Di Paolo A, Giofre C, Caglioti C, Leuzzi G, Siniscalchi A, et al. . Pharmacokinetic drug-drug interaction and their implication in clinical management. J Res Med Sci. (2013) 18:601–10.
    1. Gamelin E, Delva R, Jacob J, Merrouche Y, Raoul JL, Pezet D, et al. . Individual fluorouracil dose adjustment based on pharmacokinetic follow-up compared with conventional dosage: results of a multicenter randomized trial of patients with metastatic colorectal cancer. J Clin Oncol. (2008) 26:2099–105. 10.1200/JCO.2007.13.3934
    1. Park JR, Scott JR, Stewart CF, London WB, Naranjo A, Santana VM, et al. . Pilot induction regimen incorporating pharmacokinetically guided topotecan for treatment of newly diagnosed high-risk neuroblastoma: a Children's Oncology Group study. J Clin Oncol. (2011) 29:4351–7. 10.1200/JCO.2010.34.3293
    1. van Schaik RH. CYP450 pharmacogenetics for personalizing cancer therapy. Drug Resist Updat. (2008) 11:77–98. 10.1016/j.drup.2008.03.002
    1. Barrot CC, Woillard JB, Picard N. Big data in pharmacogenomics: current applications, perspectives and pitfalls. Pharmacogenomics. (2019) 20:609–20. 10.2217/pgs-2018-0184
    1. Hekler EB, Klasnja P, Chevance G, Golaszewski NM, Lewis D, Sim I. Why we need a small data paradigm. BMC Med. (2019) 17:133. 10.1186/s12916-019-1366-x
    1. Wong PK, Yu F, Shahangian A, Cheng G, Sun R, Ho CM. Closed-loop control of cellular functions using combinatory drugs guided by a stochastic search algorithm. Proc Natl Acad Sci USA. (2008) 105:5105–10. 10.1073/pnas.0800823105
    1. Tsutsui H, Valamehr B, Hindoyan A, Qiao R, Ding X, Guo S, et al. . An optimized small molecule inhibitor cocktail supports long-term maintenance of human embryonic stem cells. Nat Commun. (2011) 2:167. 10.1038/ncomms1165
    1. Weiss A, Ding X, van Beijnum JR, Wong I, Wong TJ, Berndsen RH, et al. . Rapid optimization of drug combinations for the optimal angiostatic treatment of cancer. Angiogenesis. (2015) 18:233–44. 10.1007/s10456-015-9462-9
    1. Mohd Abdul Rashid MB, Toh TB, Silva A, Nurrul Abdullah L, Ho CM, Ho D, et al. . Identification and optimization of combinatorial glucose metabolism inhibitors in hepatocellular carcinomas. J Lab Autom. (2015) 20:423–37. 10.1177/2211068215579612
    1. Al-Shyoukh I, Yu F, Feng J, Yan K, Dubinett S, Ho CM, et al. . Systematic quantitative characterization of cellular responses induced by multiple signals. BMC Syst Biol. (2011) 5:88. 10.1186/1752-0509-5-88
    1. Wang H, Lee DK, Chen KY, Chen JY, Zhang K, Silva A, et al. . Mechanism-independent optimization of combinatorial nanodiamond and unmodified drug delivery using a phenotypically driven platform technology. ACS Nano. (2015) 9:3332–44. 10.1021/acsnano.5b00638
    1. Pantuck AJ, Lee D-K, Kee T, Wang P, Lakhotia S, Silverman MH, et al. . Modulating BET bromodomain inhibitor ZEN-3694 and enzalutamide combination dosing in a metastatic prostate cancer patient using , an artificial intelligence platform. Adv Therapeut. (2018) 1:1800104. 10.1002/adtp.201800104
    1. Zarrinpar A, Lee DK, Silva A, Datta N, Kee T, Eriksen C, et al. . Individualizing liver transplant immunosuppression using a phenotypic personalized medicine platform. Sci Transl Med. (2016) 8:333ra49. 10.1126/scitranslmed.aac5954
    1. Blasiak A, Khong J, Kee T. : optimizing personalized medicine with artificial intelligence. SLAS Technol. (2020) 25:95–105. 10.1177/2472630319890316
    1. Schwartz LH, Litiere S, de Vries E, Ford R, Gwyther S, Mandrekar S, et al. . RECIST 1.1-update and clarification: from the RECIST committee. Eur J Cancer. (2016) 62:132–7. 10.1016/j.ejca.2016.03.081
    1. Vukobrat-Bijedic Z, Husic-Selimovic A, Sofic A, Bijedic N, Bjelogrlic I, Gogov B, et al. . Cancer antigens (CEA and CA 19-9) as markers of advanced stage of colorectal carcinoma. Med Arch. (2013) 67:397–401. 10.5455/medarh.2013.67.397-401
    1. Jing JX, Wang Y, Xu XQ, Sun T, Tian BG, Du LL, et al. . Tumor markers for diagnosis, monitoring of recurrence and prognosis in patients with upper gastrointestinal tract cancer. Asian Pac J Cancer Prev. (2014) 15:10267–72. 10.7314/APJCP.2014.15.23.10267
    1. Daamen LA, Groot VP, Heerkens HD, Intven MPW, van Santvoort HC, Molenaar IQ. Systematic review on the role of serum tumor markers in the detection of recurrent pancreatic cancer. HPB. (2018) 20:297–304. 10.1016/j.hpb.2017.11.009
    1. Kim G, Jung EJ, Ryu CG, Hwang DY. Usefulness of carcinoembryonic antigen for monitoring tumor progression during palliative chemotherapy in metastatic colorectal cancer. Yonsei Med J. (2013) 54:116–22. 10.3349/ymj.2013.54.1.116
    1. Reece M, Saluja H, Hollington P, Karapetis CS, Vatandoust S, Young GP, et al. . The use of circulating tumor DNA to monitor and predict response to treatment in colorectal cancer. Front Genet. (2019) 10:1118. 10.3389/fgene.2019.01118
    1. Parikh AR, Mojtahed A, Schneider JL, Kanter K, Van Seventer EE, Fetter IJ, et al. . Serial ctDNA monitoring to predict response to systemic therapy in metastatic gastrointestinal cancers. Clin Cancer Res. (2020) 26:1877–85. 10.1158/1078-0432.CCR-19-3467
    1. Eldridge SM, Chan CL, Campbell MJ, Bond CM, Hopewell S, Thabane L, et al. . CONSORT 2010 statement: extension to randomised pilot and feasibility trials. BMJ. (2016) 355:i5239. 10.1136/bmj.i5239
    1. Parkin DM. Global cancer statistics in the year 2000. Lancet Oncol. (2001) 2:533–43. 10.1016/S1470-2045(01)00486-7
    1. Sharma S. Tumor markers in clinical practice: general principles and guidelines. Indian J Med Paediatr Oncol. (2009) 30:1–8. 10.4103/0971-5851.56328
    1. Gennatas C, Michalaki V, Gennatas S. The role of capecitabine in the management of tumors of the digestive system. Rev Recent Clin Trials. (2009) 4:1–11. 10.2174/157488709787047576
    1. Leicher LW, de Graaf JC, Coers W, Tascilar M, de Groot JW. Tolerability of capecitabine monotherapy in metastatic colorectal cancer: a real-world study. Drugs R D. (2017) 17:117–24. 10.1007/s40268-016-0154-8
    1. Midgley R, Kerr DJ. Capecitabine: have we got the dose right? Nat Clin Pract Oncol. (2009) 6:17–24. 10.1038/ncponc1240
    1. Hirsch BR, Zafar SY. Capecitabine in the management of colorectal cancer. Cancer Manag Res. (2011) 3:79–89. 10.2147/CMAR.S11250
    1. Haller DG, Cassidy J, Clarke SJ, Cunningham D, Van Cutsem E, Hoff PM, et al. . Potential regional differences for the tolerability profiles of fluoropyrimidines. J Clin Oncol. (2008) 26:2118–23. 10.1200/JCO.2007.15.2090
    1. Diaz-Rubio E, Evans TR, Tabemero J, Cassidy J, Sastre J, Eatock M, et al. . Capecitabine (Xeloda) in combination with oxaliplatin: a phase I, dose-escalation study in patients with advanced or metastatic solid tumors. Ann Oncol. (2002) 13:558–65. 10.1093/annonc/mdf065
    1. Patt YZ, Lee FC, Liebmann JE, Diamandidis D, Eckhardt SG, Javle M, et al. . Capecitabine plus 3-weekly irinotecan (XELIRI regimen) as first-line chemotherapy for metastatic colorectal cancer: phase II trial results. Am J Clin Oncol. (2007) 30:350–7. 10.1097/COC.0b013e31804b40bb
    1. He Y, Liu P, Zhang Y, Deng X, Meng W, Wei M, et al. . Low-dose capecitabine adjuvant chemotherapy in elderly stage II/III colorectal cancer patients (LC-ACEC): study protocol for a randomized controlled trial. Trials. (2015) 16:238. 10.1186/s13063-015-0753-7

Source: PubMed

3
Abonnieren