Publications

Featured Publications

Srimongkol A, Laosillapacharoen N, Saengwimol D, Chaitankar V, Rojanaporn D, Thanomchard T, Borwornpinyo S, Hongeng S, and Kaewkhaw R. J Exp Clin Cancer Res. 2023 Feb 1;42(1):39.
doi: 10.1186/s13046-023-02608-1. PMID: 36726110; PMCID: PMC9890748. (Q1 (top 10), Impact factor: 12.7)

Sunitinib efficacy with minimal toxicity in patient-derived retinoblastoma organoids

Background

Recurrence of retinoblastoma (RB) following chemoreduction is common and is often managed with local (intra-arterial/intravitreal) chemotherapy. However, some tumors are resistant to even local administration of maximum feasible drug dosages, or effective tumor control and globe preservation may be achieved at the cost of vision loss due to drug-induced retinal toxicity. The aim of this study was to identify drugs with improved antitumor activity and more favorable retinal toxicity profiles via screening of potentially repurposable FDA-approved drugs in patient-derived tumor organoids.

Methods

Genomic profiling of five RB organoids and the corresponding parental tissues was performed. RB organoids were screened with 133 FDA-approved drugs, and candidate drugs were selected based on cytotoxicity and potency. RNA sequencing was conducted to generate a drug signature from RB organoids, and the effects of drugs on cell cycle progression and proliferative tumor cone restriction were examined. Drug toxicity was assessed with human embryonic stem cell-derived normal retinal organoids. The efficacy/toxicity profiles of candidate drugs were compared with those of drugs in clinical use.

Results

RB organoids maintained the genomic features of the parental tumors. Sunitinib was identified as highly cytotoxic against both classical RB1-deficient and novel MYCN-amplified RB organoids and inhibited proliferation while inducing differentiation in RB. Sunitinib was a more effective suppressor of proliferative tumor cones in RB organoids and had lower toxicity in normal retinal organoids than either melphalan or topotecan.

Conclusion

The efficacy and retinal toxicity profiles of sunitinib suggest that it could potentially be repurposed for local chemotherapy of RB.


Chittavanich P, Saengwimol D, Roytrakul S, Rojanaporn D, Chaitankar V, Srimongkol A, Anurathapan U, Hongeng S, Kaewkhaw R. Mol Oncol. 2023 Nov 17.
doi: 10.1002/1878-0261.13553. Epub ahead of print. PMID: 37975412. (Q1 (top 10), Impact factor: 7.5)

Ceftriaxone exerts antitumor effects in MYCN-driven retinoblastoma and neuroblastoma by targeting DDX3X for translation repression

MYCN proto-oncogene, bHLH transcription factor (MYCN) amplification is associated with aggressive retinoblastoma (RB) and neuroblastoma (NB) cancer recurrence that is resistant to chemotherapies. Therefore, there is an urgent need to identify new therapeutic tools. This study aimed to evaluate the potential repurposing of ceftriaxone for the treatment of MYCN-amplified RB and NB, based on the clinical observations that the drug was serendipitously found to decrease the volume of the MYCN-driven RB subtype. Using patient-derived tumor organoids and tumor cell lines, we demonstrated that ceftriaxone is a potent and selective growth inhibitor targeting MYCN-driven RB and NB cells. Profiling of drug-induced transcriptomic changes, cell-cycle progression, and apoptotic death indicated cell-cycle arrest and death of drug-treated MYCN-amplified tumor cells. Drug target identification, using an affinity-based proteomic and molecular docking approach, and functional studies of the target proteins revealed that ceftriaxone targeted DEAD-box helicase 3 X-linked (DDX3X), thereby inhibiting translation in MYCN-amplified tumors but not in MYCN-nonamplified cells. The data suggest the feasibility of repurposing ceftriaxone as an anticancer drug and provide insights into the mechanism of drug action, highlighting DDX3X as a potential target for treating MYCN-driven tumors.


Publications

2022

Rojanaporn D, Chitphuk S, Iemwimangsa N, Chareonsirisuthigul T, Saengwimol D, Aroonroch R, Anurathathapan U, Hongeng S, Kaewkhaw R. Germline RB1 Mutation in Retinoblastoma Patients: Detection Methods and Implication in Tumor Focality. Translational Vision Science and Technology, 2022, 11(9), 30. (Q1, Impact factor: 3.05)
doi: 10.1167/tvst.11.9.30

2021

Pairoj S, Damrongsak P, Damrongsak B, Jinawath N, Kaewkhaw R, Ruttanasirawit C, Leelawattananon T, Locharoenrat K. Antitumor activities of carboplatin-doxorubicin-ZnO complexes in different human cancer cell lines (breast, cervix uteri, colon, liver and oral) under UV exposition. Artificial Cells, Nanomedicine and Biotechnology, 2021, 49(1), pp. 120–135. (Q1)
doi: 10.1080/21691401.2021.1876718

2020

Saengwimol D, Chittavanich P, Laosillapacharoen N, Srimongkol A, Chaitankar V, Rojanaporn D, Aroonroch R, Suktitipat B, Saisawang C, Svasti S, Hongeng S, Kaewkhaw R. Silencing of the Long Noncoding RNA MYCNOS1 Suppresses Activity of MYCN-Amplified Retinoblastoma Without RB1 Mutation. Invest Ophthalmol Vis Sci. 2020 Dec 1;61(14):8.
doi: 10.1167/iovs.61.14.8. PMID: 33270844; PMCID: PMC7718827. (Q1 (top10), Impact factor: 4.93)

Kaewkhaw R, Rojanaporn D. Retinoblastoma: Etiology, Modeling, and Treatment. Cancers (Basel). 2020 Aug 16;12(8):2304.
doi: 10.3390/cancers12082304. PMID: 32824373; PMCID: PMC7465685.( Q1, Impact factor: 6.58)

2019

Pairoj, S., Damrongsak, P., Damrongsak, B., Jinawath, N., Kaewkhaw, R., Leelawattananon, T., Ruttanasirawit, C, Locharoenrat, K. Antiradical properties of chemo drug, carboplatin, in cooperation with ZnO nanoparticles under UV irradiation in putative model of cancer cells. Biocybernetics and Biomedical Engineering. 2019, 39(3), 893–901.
doi: 10.1016/j.bbe.2019.08.004. (Q2)

2018

Saengwimol D, Rojanaporn D, Chaitankar V, Chittavanich P, Aroonroch R, Boontawon T, Thammachote W, Jinawath N, Hongeng S, Kaewkhaw R. A three-dimensional organoid model recapitulates tumorigenic aspects and drug responses of advanced human retinoblastoma. Sci Rep. 2018 Oct 23;8(1):15664.
doi: 10.1038/s41598-018-34037-y. PMID: 30353124; PMCID: PMC6199308. (Q1, (top 10), Impact factor: 4.99)

Rojanaporn D, Boontawon T, Chareonsirisuthigul T, Thanapanpanich O, Attaseth T, Saengwimol D, Anurathapan U, Sujirakul T, Kaewkhaw R*, Hongeng S. Spectrum of germline RB1 mutations and clinical manifestations in retinoblastoma patients from Thailand. Mol Vis. 2018 Dec 9; 24:778-788. eCollection 2018. PubMed PMID: 30636860; PubMed Central PMCID: PMC6300611. (Q2, Impact factor: 2.71) 

2016

Kaewkhaw R, Swaroop M, Homma K, Nakamura J, Brooks M, Kaya KD, Chaitankar V, Michael S, Tawa G, Zou J, Rao M, Zheng W, Cogliati T, Swaroop A. Treatment paradigms for retinal and macular diseases using 3-D retina cultures derived from human reporter pluripotent stem cell lines. Invest Ophthalmol Vis Sci. 2016 Apr 1;57(5): ORSFl1-ORSFl11.
doi: 10.1167/iovs.15-17639. PubMed PMID: 27116668; PubMed Central PMCID: PMC4855830. (Q1, Impact factor: 4.93)

2015

Kaewkhaw R, Kaya KD, Brooks M, Homma K, Zou J, Chaitankar V, Rao M, Swaroop A.Transcriptome dynamics of developing photoreceptors in three-dimensional retina cultures recapitulates temporal sequence of human cone and rod differentiation revealing cell surface markers and gene networks. Stem Cells. 2015 Dec;33(12):3504-18.
doi: 10.1002/stem.2122. Epub 2015 Aug 14. PubMed PMID: 26235913; PubMed Central PMCID: PMC4713319. (Q1, Impact factor: 6.52)

2012

Kaewkhaw R, Scutt AM, Haycock JW. Integrated culture and purification of rat Schwann cells from freshly isolated adult tissue. Nat Protoc. 2012 Nov;7(11):1996-2004.
doi: 10.1038/nprot.2012.118. Epub 2012 Oct 11. PubMed PMID: 23060244. (Q1 (top 10), Impact factor: 10.419)

2011

Kaewkhaw R, Scutt AM, Haycock JW. Anatomical site influences the differentiation of adipose-derived stem cells for Schwann-cell phenotype and function. Glia. 2011 May;59(5):734-49.
doi: 10.1002/glia.21145. Epub 2011 Feb 23. PubMed PMID: 21351157. (Q1 (top 10), Impact factor: 5.984)