A nanoengineered topical transmucosal cisplatin delivery system induces anti-tumor response in animal models and patients with oral cancer

Manijeh Goldberg, Aaron Manzi, Amritpreet Birdi, Brandon Laporte, Peter Conway, Stefanie Cantin, Vasudha Mishra, Alka Singh, Alexander T Pearson, Eric R Goldberg, Sam Goldberger, Benjamin Flaum, Rifat Hasina, Nyall R London, Gary L Gallia, Chetan Bettegowda, Simon Young, Vlad Sandulache, James Melville, Jonathan Shum, Sonya E O'Neill, Erkin Aydin, Alex Zhavoronkov, Anxo Vidal, Atenea Soto, Maria Jose Alonso, Ari J Rosenberg, Mark W Lingen, Anil D'Cruz, Nishant Agrawal, Evgeny Izumchenko, Manijeh Goldberg, Aaron Manzi, Amritpreet Birdi, Brandon Laporte, Peter Conway, Stefanie Cantin, Vasudha Mishra, Alka Singh, Alexander T Pearson, Eric R Goldberg, Sam Goldberger, Benjamin Flaum, Rifat Hasina, Nyall R London, Gary L Gallia, Chetan Bettegowda, Simon Young, Vlad Sandulache, James Melville, Jonathan Shum, Sonya E O'Neill, Erkin Aydin, Alex Zhavoronkov, Anxo Vidal, Atenea Soto, Maria Jose Alonso, Ari J Rosenberg, Mark W Lingen, Anil D'Cruz, Nishant Agrawal, Evgeny Izumchenko

Abstract

Despite therapeutic advancements, oral cavity squamous cell carcinoma (OCSCC) remains a difficult disease to treat. Systemic platinum-based chemotherapy often leads to dose-limiting toxicity (DLT), affecting quality of life. PRV111 is a nanotechnology-based system for local delivery of cisplatin loaded chitosan particles, that penetrate tumor tissue and lymphatic channels while avoiding systemic circulation and toxicity. Here we evaluate PRV111 using animal models of oral cancer, followed by a clinical trial in patients with OCSCC. In vivo, PRV111 results in elevated cisplatin retention in tumors and negligible systemic levels, compared to the intravenous, intraperitoneal or intratumoral delivery. Furthermore, PRV111 produces robust anti-tumor responses in subcutaneous and orthotopic cancer models and results in complete regression of carcinogen-induced premalignant lesions. In a phase 1/2, open-label, single-arm trial (NCT03502148), primary endpoints of efficacy (≥30% tumor volume reduction) and safety (incidence of DLTs) of neoadjuvant PRV111 were reached, with 69% tumor reduction in ~7 days and over 87% response rate. Secondary endpoints (cisplatin biodistribution, loco-regional control, and technical success) were achieved. No DLTs or drug-related serious adverse events were reported. No locoregional recurrences were evident in 6 months. Integration of PRV111 with current standard of care may improve health outcomes and survival of patients with OCSCC.

Conflict of interest statement

M.G., A.M., A.B., B.L., P.C., S.C., B.F. and E.R.G. are affiliated with Privo Technologies. N.A. and S.G. serve as advisors for Privo Technologies. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Cisplatin chitosan nanoparticles (CDDP-NP) demonstrate…
Fig. 1. Cisplatin chitosan nanoparticles (CDDP-NP) demonstrate improved local drug retention and induce potent anti-tumor response in FaDu tumor bearing mice xenografts.
A Schematic representation of the experiment. B Nude mice xenografts bearing subcutaneous human FaDu HNSCC cell line induced tumors were treated with either PBS-IT, CDDP-IT, CDDP-IV, BLK-NP, or CDDP-NP. Graphs show the average tumor volume for 6 animals per group ± SEM. P-value was performed by a 1-sided Wilcoxon Rank-sum Test. Asterisk represents statistical significance between the CDDP-IT and CDDP-NP groups (p < 0.05). C Residual tumors were harvested after 4 treatments (day 14) for CDDP-NP (n = 6) or CDDP-IT (n = 4) and 2 treatments (day 7) for CDDP-IV (n = 4). Samples were weighted, homogenized and cisplatin level was quantified by ICP-MS, shown as average ± SEM (p < 0.0001; Student’s unpaired t-test). D Blood was collected from 4 animals per group after administration of the first dose at indicated time points and level of cisplatin was quantified by ICP-MS, shown as average ± SEM. E Graph shows the average body weights for 6 animals per group ± SEM. Source data are provided as a Source Data file.
Fig. 2. Local application of PRV111 induced…
Fig. 2. Local application of PRV111 induced robust anti-tumor response in hamster orthotopic oral cancer model.
A Schematic representation of the experiment. B Golden Syrian hamsters bearing orthotopic tumors induced by injection of HCPC-1 cell line into the cheek pouch were treated with either PBS-IP, CDDP-IP, BLK-patch or PRV111. Graphs show the average tumor volume for 6 animals per group ± SEM. P-value was performed by a 1-sided Wilcoxon Rank-sum method. Asterisk represents statistical significance between the CDDP-IP and PRV111 groups (p < 0.05). C Graph shows the average body weights for 6 animals per group ± SEM. Body weights of 5 tumor free animals without treatment were measure along the tumor bearing counterparts (dashed line). D Representative fluorescence images of tumor sections at indicated magnification taken after treatment of the hamster with PRV111 patch containing chitosan particles labeled with Cy5 (red) and encapsulating FITC (green). Yellow areas display dual-labeling, NPs with encapsulated FITC. Permeation experiment was repeated in 6 tumors. Source data are provided as a Source Data file.
Fig. 3. Topical administration of PRV111 reduced…
Fig. 3. Topical administration of PRV111 reduced cisplatin associated toxicities and prevented tumor recurrence in vivo.
A Blood was collected after administration of the first dose of CDDP-IP or PRV111 (6 animals per group), and level of cisplatin was quantified by ICP-MS, shown as average ± SEM (p = 0.0001; Student’s unpaired t-test). B, C Hamsters treated with CDDP-IP or PRV111 (6 animals per group) were sacrificed and residual tumors as well as lungs, heart, brain, liver, kidneys, stomach, spleen, healthy contralateral cheek pouch, and tongue were harvested. Samples were weighted, processed, and biodistribution of cisplatin was assessed by ICP-MS, shown as average ± SEM (p = 0.005 for tumor tissue all body organs analyzed; Student’s unpaired t-test). Note, residual tumors were collected from four animals treated with PRV111. D Representative H&E stained histopathological images of kidneys (×200; scale bar—100 μm). Three tumors per group were stained. E Animals that were tumor free after treatment with either PRV111 or CDDP-IP (6 per group) were monitored for recurrence and survival during the indicated time period (months) post treatment cessation. Animals were evaluated and weighted weekly until day 45, and once a month thereafter. Cyan squares indicate a month when animal remained tumor-free. Pink squares indicate a month when recurrence was detected. Tumor volume upon recurrence is provided next to the each pink square. Source data are provided as a Source Data file.
Fig. 4. PRV111 induced rapid and sustainable…
Fig. 4. PRV111 induced rapid and sustainable regression of carcinogen-induced oral premalignant lesions.
A Schematic representation of the experiment. B Golden Syrian hamsters bearing DMBA-induced oral dysplastic lesion were treated with either BLK-patch, CDDP-IP or PRV111 (3 animals per group). Red circle indicates presence of the premalignant lesion, whereas blue circle indicates that the premalignant lesions was undetected. C Graph shows the average body weights for the 3 hamsters in each indicated group  ± SEM. D Blood was collected from 3 animals per group after administration of the first dose of CDDP-IP or PRV111, and level of cisplatin was quantified by ICP-MS, shown as average ± SEM (p < 0.0019; Student’s unpaired t-test). E Hamsters treated with CDDP-IP or PRV111 (n = 3) were sacrificed and indicated organs were harvested, weighted, processed, and biodistribution of cisplatin was assessed by ICP-MS, shown as average ± SEM (p < 0.05 for all body organs analyzed; Student’s unpaired t-test). F Representative H&E stained histopathological images of kidneys (200×; scale bar—100 μm). Three tumors per group were stained. Source data are provided as a Source Data file.
Fig. 5. PRV111 induced a rapid anti-tumor…
Fig. 5. PRV111 induced a rapid anti-tumor response in patients with locally advanced OCSCC.
A Schematic representation of the clinical trial timeline. B Tumor volume changes from baseline and end of study for each patient treated with PRV111. C Tumor photographs and volume measurements for subjects which achieved >90% tumor volume reduction post RPV111 treatment. D Representative H&E image of post-PRV111 treatment tumor sample. Top: Background of residual cancer cells (black circles) and evidence of treatment effect in the form of necrosis/ulceration, cellular debris, and acute/chronic inflammation (blue circles). 4×; scale bar – 1000um. Bottom: Area of minimal residual cancer cells and evidence of treatment effect in the form of necrosis/ulceration and cellular debris, as indicated by black arrows. 40×; scale bar—100μm. E Biodistribution of platinum detected in the blood, and tumor tissue following PRV111 therapy compared with standard of care cisplatin treatment.
Fig. 6. PRV111 increased the number of…
Fig. 6. PRV111 increased the number of TILs.
A Fold change increase in TILs after treatment of 8 patients with PRV111 relative to pretreatment biopsies, shown as average ± SEM (p < 0.001; Student’s unpaired t-test). Four slides were analyzed for each specimen and average was used for fold change calculation. B Representative images (10×; scale bar—1000μm) of pre- and post-treatment sections stained for CD3, CD8, and CD4 T cells. Changes in brown staining indicates increase in tumor-infiltrating lymphocytes following PRV111 treatment. Source data are provided as a Source Data file.

References

    1. Howlader N, N. A. et al. (eds). SEER Cancer Statistics Review, 1975-2017, National Cancer Institute. Bethesda, MD, , based on November 2019 SEER data submission, posted to the SEER web site, April 2020. (2020).
    1. Kramer S, et al. Combined radiation-therapy and surgery in the management of advanced head and neck-cancer - final report of study 73-03 of the radiation-therapy-oncology-group. Head. Neck Surg. 1987;10:19–30. doi: 10.1002/hed.2890100105.
    1. Brizel DM, et al. Hyperfractionated irradiation with or without concurrent chemotherapy for locally advanced head and neck cancer. N. Engl. J. Med. 1998;338:1798–1804. doi: 10.1056/NEJM199806183382503.
    1. Cohen EE, Lingen MW, Vokes EE. The expanding role of systemic therapy in head and neck cancer. J. Clin. Oncol. 2004;22:1743–1752. doi: 10.1200/JCO.2004.06.147.
    1. Pignon JP, Bourhis J, Domenge C, Designe L, Grp MNC. Chemotherapy added to locoregional treatment for head and neck squamous-cell carcinoma: three meta-analyses of updated individual data. Lancet. 2000;355:949–955. doi: 10.1016/S0140-6736(00)90011-4.
    1. Wang TJ, et al. Influences of facial disfigurement and social support for psychosocial adjustment among patients with oral cancer in Taiwan: a cross-sectional study. BMJ Open. 2018;8:e023670. doi: 10.1136/bmjopen-2018-023670.
    1. Kam D, et al. Incidence of suicide in patients with head and neck cancer. JAMA Otolaryngol. Head. Neck Surg. 2015;141:1075–1081. doi: 10.1001/jamaoto.2015.2480.
    1. Johnson DE, et al. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Prim. 2020;6:92. doi: 10.1038/s41572-020-00224-3.
    1. Machiels JP, et al. Squamous cell carcinoma of the oral cavity, larynx, oropharynx and hypopharynx: EHNS–ESMO–ESTRO Clinical Practice Guidelines for diagnosis, treatment and follow-up††These Guidelines were developed by the European Head and Neck Society (EHNS), the European Society for Medical Oncology (ESMO) and the European SocieTy for Radiotherapy & Oncology (ESTRO). The three societies nominated authors to write the guidelines as well as reviewers to comment on them. These guidelines were approved by the EHNS Executive Board, the ESMO Guidelines Committee and the ESTRO Executive Board in June 2020. Ann. Oncol. 2020;31:1462–1475. doi: 10.1016/j.annonc.2020.07.011.
    1. Cepeda V, et al. Biochemical mechanisms of cisplatin cytotoxicity. Anti-cancer agents medicinal Chem. 2007;7:3–18. doi: 10.2174/187152007779314044.
    1. Magrini SM, et al. Cetuximab and radiotherapy versus cisplatin and radiotherapy for locally advanced head and neck cancer: a randomized phase II trial. J. Clin. Oncol. 2016;34:427–435. doi: 10.1200/JCO.2015.63.1671.
    1. Lee Y-G, et al. Treatment strategy and outcomes in locally advanced head and neck squamous cell carcinoma: a nationwide retrospective cohort study (KCSG HN13–01) BMC Cancer. 2020;20:813. doi: 10.1186/s12885-020-07297-z.
    1. Holleran WM, DeGregorio MW. Evolution of high-dose cisplatin. Investigational N. Drugs. 1988;6:135–142.
    1. Duan X, He C, Kron SJ, Lin W. Nanoparticle formulations of cisplatin for cancer therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2016;8:776–791. doi: 10.1002/wnan.1390.
    1. Boztepe T, Castro GR, León IE. Lipid, polymeric, inorganic-based drug delivery applications for platinum-based anticancer drugs. Int J. Pharm. 2021;605:120788. doi: 10.1016/j.ijpharm.2021.120788.
    1. Farooq MA, et al. Recent progress in nanotechnology-based novel drug delivery systems in designing of cisplatin for cancer therapy: an overview. Artif. Cells Nanomed. Biotechnol. 2019;47:1674–1692. doi: 10.1080/21691401.2019.1604535.
    1. Goldberg M, et al. Development of a nanoparticle-embedded chitosan sponge for topical and local administration of chemotherapeutic agents. J. Nanotechnol. Eng. Med. 2014;5:0409051–4090511. doi: 10.1115/1.4030899.
    1. Miyano K, et al. cRGD peptide installation on cisplatin-loaded nanomedicines enhances efficacy against locally advanced head and neck squamous cell carcinoma bearing cancer stem-like cells. J. Controlled Release. 2017;261:275–286. doi: 10.1016/j.jconrel.2017.06.021.
    1. Davidi ES, et al. Cisplatin-conjugated gold nanoparticles as a theranostic agent for head and neck cancer. Head. Neck. 2018;40:70–78. doi: 10.1002/hed.24935.
    1. Cohen SM, et al. Efficacy and toxicity of peritumoral delivery of nanoconjugated cisplatin in an in vivo murine model of head and neck squamous cell carcinoma. JAMA Otolaryngol. Head. Neck Surg. 2013;139:382–387. doi: 10.1001/jamaoto.2013.214.
    1. Endo K, et al. Tumor-targeted chemotherapy with the nanopolymer-based drug NC-6004 for oral squamous cell carcinoma. Cancer Sci. 2013;104:369–374. doi: 10.1111/cas.12079.
    1. Kim JH, et al. Antitumor efficacy of cisplatin-loaded glycol chitosan nanoparticles in tumor-bearing mice. J. Controlled Release Off. J. Controlled Release. Soc. 2008;127:41–49. doi: 10.1016/j.jconrel.2007.12.014.
    1. Lin J, Alexander-Katz A. Cell membranes open “doors” for cationic nanoparticles/biomolecules: insights into uptake kinetics. ACS Nano. 2013;7:10799–10808. doi: 10.1021/nn4040553.
    1. Sarin H. Physiologic upper limits of pore size of different blood capillary types and another perspective on the dual pore theory of microvascular permeability. J. Angiogenes Res. 2010;2:14. doi: 10.1186/2040-2384-2-14.
    1. Ferreira MBA, Lima JPSN, Cohen EEW. Novel targeted therapies in head and neck cancer. Expert Opin. Investigational Drugs. 2012;21:281–295. doi: 10.1517/13543784.2012.651455.
    1. Odukoya O, Schwartz J, Weichselbaum R, Shklar G. An epidermoid carcinoma cell line derived from hamster 7,12- dimethylbenz[a]anthracene-induced buccal pouch tumors2. JNCI: J. Natl Cancer Inst. 1983;71:1253–1264.
    1. Vidya Priyadarsini R, et al. Gene expression signature of DMBA-induced hamster buccal pouch carcinomas: modulation by chlorophyllin and ellagic acid. PLoS ONE. 2012;7:e34628. doi: 10.1371/journal.pone.0034628.
    1. Chen D, Yang K, Zhang G, Mei J, Xiang L. Screen and analysis of key disease genes for precancerous lesions of oral buccal mucosa induced by DMBA in golden hamsters. Oncol. Lett. 2011;2:265–271. doi: 10.3892/ol.2010.228.
    1. Chiang CP, Huang WT, Lee JW, Hsu YC. Effective treatment of 7,12-dimethylbenz(a)anthracene-induced hamster buccal pouch precancerous lesions by topical photosan-mediated photodynamic therapy. Head. Neck. 2012;34:505–512. doi: 10.1002/hed.21761.
    1. Hsu YC, Yang DF, Chiang CP, Lee JW, Tseng MK. Successful treatment of 7,12-dimethylbenz(a)anthracene-induced hamster buccal pouch precancerous lesions by topical 5-aminolevulinic acid-mediated photodynamic therapy. Photodiagnosis Photodyn. Ther. 2012;9:310–318. doi: 10.1016/j.pdpdt.2012.03.003.
    1. Wang YY, et al. MAL-PDT inhibits oral precancerous cells and lesions via autophagic cell death. Oral. Dis. 2019;25:758–771. doi: 10.1111/odi.13036.
    1. Shen L, Xu Q, Li P, Zhou G. Efficacy of krypton laser photodynamic therapy for oral mucosa dysplasia in 9,10-dimethyl-1,2-benzanthracene-treated hamsters. Oncol. Lett. 2013;6:1358–1362. doi: 10.3892/ol.2013.1554.
    1. Chen D, et al. Screening the pathogenic genes and pathways related to DMBA (7,12-dimethylbenz[a]anthracene)-induced transformation of hamster oral mucosa from precancerous lesions to squamous cell carcinoma. Oncol. Lett. 2011;2:637–642. doi: 10.3892/ol.2011.293.
    1. Hann S, et al. Platinum determination by inductively coupled plasma-sector field mass spectrometry (ICP-SFMS) in different matrices relevant to human biomonitoring. Anal. Bioanal. Chem. 2003;376:198–204. doi: 10.1007/s00216-003-1861-x.
    1. Morrison JG, et al. Validation of a highly sensitive ICP-MS method for the determination of platinum in biofluids: application to clinical pharmacokinetic studies with oxaliplatin. J. Pharm. Biomed. Anal. 2000;24:1–10. doi: 10.1016/S0731-7085(00)00377-0.
    1. Wang, J. et al. The protein-binding behavior of platinum anticancer drugs in blood revealed by mass spectrometry. Pharmaceuticals14, 104 (2021).
    1. Martinčič A, et al. A novel method for speciation of Pt in human serum incubated with cisplatin, oxaliplatin and carboplatin by conjoint liquid chromatography on monolithic disks with UV and ICP-MS detection. Talanta. 2013;116:141–148. doi: 10.1016/j.talanta.2013.05.016.
    1. Kulkarni U, Mahalingam R, Pather SI, Li X, Jasti B. Porcine buccal mucosa as an in vitro model: relative contribution of epithelium and connective tissue as permeability barriers. J. Pharm. Sci. 2009;98:471–483. doi: 10.1002/jps.21436.
    1. Kinikoglu B, Hemar J, Hasirci V, Breton P, Damour O. Feasibility of a porcine oral mucosa equivalent: a preclinical study. Artif. Cells Blood Substit. Biotechnol. 2012;40:271–274. doi: 10.3109/10731199.2011.644293.
    1. Neville BW, Day TA. Oral cancer and precancerous lesions. CA Cancer J. Clin. 2002;52:195–215. doi: 10.3322/canjclin.52.4.195.
    1. Ribeiro AS, Salles PR, da Silva TA, Mesquita RA. A review of the nonsurgical treatment of oral leukoplakia. Int. J. Dent. 2010;2010:186018. doi: 10.1155/2010/186018.
    1. Chau L, et al. Topical agents for oral cancer chemoprevention: a systematic review of the literature. Oral. Oncol. 2017;67:153–159. doi: 10.1016/j.oraloncology.2017.02.014.
    1. Chow SC, Tu YH. On two-stage seamless adaptive design in clinical trials. J. Formos. Med Assoc. 2008;107:52–60. doi: 10.1016/S0929-6646(09)60009-7.
    1. Shan G, Wilding GE, Hutson AD, Gerstenberger S. Optimal adaptive two-stage designs for early phase II clinical trials. Stat. Med. 2016;35:1257–1266. doi: 10.1002/sim.6794.
    1. Simon R. Optimal two-stage designs for phase II clinical trials. Control Clin. Trials. 1989;10:1–10. doi: 10.1016/0197-2456(89)90015-9.
    1. Florea AM, Büsselberg D. Cisplatin as an anti-tumor drug: cellular mechanisms of activity, drug resistance and induced side effects. Cancers. 2011;3:1351–1371. doi: 10.3390/cancers3011351.
    1. Gouyette A, Apchin A, Foka M, Richard JM. Pharmacokinetics of intra-arterial and intravenous cisplatin in head and neck cancer patients. Eur. J. Cancer Clin. Oncol. 1986;22:257–263. doi: 10.1016/0277-5379(86)90389-5.
    1. Rajkumar P, et al. Cisplatin concentrations in long and short duration infusion: implications for the optimal time of radiation Delivery. J. Clin. Diagnostic Res JCDR. 2016;10:XC01–XC04.
    1. Nguyen N, et al. Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head. neck. 2016;38:1074–1084. doi: 10.1002/hed.24406.
    1. Fang J, et al. Prognostic significance of tumor infiltrating immune cells in oral squamous cell carcinoma. BMC cancer. 2017;17:375. doi: 10.1186/s12885-017-3317-2.
    1. Balermpas P, et al. Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer. Br. J. Cancer. 2014;110:501–509. doi: 10.1038/bjc.2013.640.
    1. de Biasi AR, Villena-Vargas J, Adusumilli PS. Cisplatin-induced antitumor immunomodulation: a review of preclinical and clinical evidence. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014;20:5384–5391. doi: 10.1158/1078-0432.CCR-14-1298.
    1. Wakita D, et al. Cisplatin augments antitumor T-cell responses leading to a potent therapeutic effect in combination with PD-L1 blockade. Anticancer Res. 2019;39:1749–1760. doi: 10.21873/anticanres.13281.
    1. Broner EC, et al. Doublecortin-like kinase 1 (DCLK1) is a novel NOTCH pathway signaling regulator in head and neck squamous cell carcinoma. Front Oncol. 2021;11:677051. doi: 10.3389/fonc.2021.677051.
    1. Sankar V, et al. Local drug delivery for oral mucosal diseases: challenges and opportunities. Oral. Dis. 2011;17:73–84. doi: 10.1111/j.1601-0825.2011.01793.x.
    1. Gupta V, Karthikeyan C, Trivedi P. Localized delivery of cisplatin for the effective management of squamous cell carcinoma from protransfersome formulation. Arch. Pharm. Res. 2012;35:851–859. doi: 10.1007/s12272-012-0510-3.
    1. Sudha T, et al. Targeted delivery of cisplatin to tumor xenografts via the nanoparticle component of nano-diamino-tetrac. Nanomed. 2017;12:195–205. doi: 10.2217/nnm-2016-0315.
    1. Wenig BL, et al. The role of intratumoral therapy with cisplatin/epinephrine injectable gel in the management of advanced squamous cell carcinoma of the head and neck. Arch. Otolaryngol. Head. Neck Surg. 2002;128:880–885. doi: 10.1001/archotol.128.8.880.
    1. Li J, et al. Biodegradable thermosensitive hydrogel for SAHA and DDP delivery: therapeutic effects on oral squamous cell carcinoma xenografts. PLoS ONE. 2012;7:e33860. doi: 10.1371/journal.pone.0033860.
    1. Desai KGH. Polymeric drug delivery systems for intraoral site-specific chemoprevention of oral cancer. J. Biomed. Mater. Res B Appl Biomater. 2018;106:1383–1413. doi: 10.1002/jbm.b.33943.
    1. Hu D, et al. A novel modular polymer platform for the treatment of head and neck squamous cell carcinoma in an animal model. Arch. Otolaryngol.–Head. Neck Surg. 2012;138:412–417. doi: 10.1001/archoto.2012.20.
    1. Lan X, et al. Microneedle-mediated delivery of lipid-coated cisplatin nanoparticles for efficient and safe cancer therapy. ACS Appl. Mater. interfaces. 2018;10:33060–33069. doi: 10.1021/acsami.8b12926.
    1. Cai S, Xie Y, Davies NM, Cohen MS, Forrest ML. Carrier-based intralymphatic cisplatin chemotherapy for the treatment of metastatic squamous cell carcinoma of the head & neck. Therapeutic Deliv. 2010;1:237–245. doi: 10.4155/tde.10.38.
    1. Osada A, et al. 132 Poster - Phase IIa/IIb clinical trial of NC-6004 (Nanoparticle Cisplatin) plus Pembrolizumab in patients with head and neck cancer (HNSCC) who have failed platinum or a platinum-containing regimen. Eur. J. Cancer. 2020;138:S35. doi: 10.1016/S0959-8049(20)31164-3.
    1. Agrawal N, et al. A phase II study of PRV111 nanoengineered cisplatin patch as a neoadjuvant therapy for early-stage oral squamous cell carcinoma (OSCC) J. Clin. Oncol. 2021;39:6056–6056. doi: 10.1200/JCO.2021.39.15_suppl.6056.
    1. Calixto G, Bernegossi J, Fonseca-Santos B, Chorilli M. Nanotechnology-based drug delivery systems for treatment of oral cancer: a review. Int. J. Nanomed. 2014;9:3719–3735. doi: 10.2147/IJN.S61670.
    1. Zhang M, et al. Current trends of targeted drug delivery for oral cancer therapy. Front. Bioeng. Biotechnol. 2020;8:618931. doi: 10.3389/fbioe.2020.618931.
    1. Quiñones, J. P., Peniche, H. & Peniche, C. Chitosan based self-assembled nanoparticles in drug delivery. Polymers (Basel)10 (2018).
    1. Delan WK, et al. Formulation of simvastatin chitosan nanoparticles for controlled delivery in bone regeneration: optimization using Box-Behnken design, stability and in vivo study. Int. J. Pharmaceutics. 2020;577:119038. doi: 10.1016/j.ijpharm.2020.119038.
    1. Duceppe N, Tabrizian M. Advances in using chitosan-based nanoparticles for in vitro and in vivo drug and gene delivery. Expert Opin. Drug Deliv. 2010;7:1191–1207. doi: 10.1517/17425247.2010.514604.
    1. Elgadir MA, et al. Impact of chitosan composites and chitosan nanoparticle composites on various drug delivery systems: a review. J. Food Drug Anal. 2015;23:619–629. doi: 10.1016/j.jfda.2014.10.008.
    1. Wang JJ, et al. Recent advances of chitosan nanoparticles as drug carriers. Int J. Nanomed. 2011;6:765–774.
    1. Remuñán-López C, Lorenzo-Lamosa ML, Vila-Jato JL, Alonso MJ. Development of new chitosan-cellulose multicore microparticles for controlled drug delivery. Eur. J. Pharm. Biopharm. 1998;45:49–56. doi: 10.1016/S0939-6411(97)00122-7.
    1. Mazzarino L, et al. Curcumin-loaded chitosan-coated nanoparticles as a new approach for the local treatment of oral cavity cancer. J. Nanosci. Nanotechnol. 2015;15:781–791. doi: 10.1166/jnn.2015.9189.
    1. Costa Idos S, Abranches RP, Garcia MT, Pierre MB. Chitosan-based mucoadhesive films containing 5-aminolevulinic acid for buccal cancer’s treatment. J. Photochem Photobio. B. 2014;140:266–275. doi: 10.1016/j.jphotobiol.2014.08.005.
    1. Remuñán-López C, Portero A, Vila-Jato JL, Alonso MJ. Design and evaluation of chitosan/ethylcellulose mucoadhesive bilayered devices for buccal drug delivery. J. Control Release. 1998;55:143–152. doi: 10.1016/S0168-3659(98)00044-3.
    1. Staffhorst RW, et al. Antitumor activity and biodistribution of cisplatin nanocapsules in nude mice bearing human ovarian carcinoma xenografts. Anti-cancer drugs. 2008;19:721–727. doi: 10.1097/CAD.0b013e328304355f.
    1. Alsmadi MM, Obaidat RM, Alnaief M, Albiss BA, Hailat N. Development, in vitro characterization, and in vivo toxicity evaluation of chitosan-alginate nanoporous carriers loaded with cisplatin for lung cancer treatment. AAPS PharmSciTech. 2020;21:191. doi: 10.1208/s12249-020-01735-8.
    1. Fröhlich E. The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles. Int. J. Nanomed. 2012;7:5577–5591. doi: 10.2147/IJN.S36111.
    1. Werner JA, et al. A phase III placebo-controlled study in advanced head and neck cancer using intratumoural cisplatin/epinephrine gel. Br. J. Cancer. 2002;87:938–944. doi: 10.1038/sj.bjc.6600588.
    1. Gimenez-Conti IB, Slaga TJ. The hamster cheek pouch carcinogenesis model. J. Cell Biochem Suppl. 1993;17f:83–90. doi: 10.1002/jcb.240531012.
    1. Shklar G. Development of experimental oral carcinogenesis and its impact on current oral cancer research. J. Dent. Res. 1999;78:1768–1772. doi: 10.1177/00220345990780120101.
    1. Manimaran A, Manoharan S. Tumor preventive efficacy of emodin in 7,12-Dimethylbenz[a]Anthracene-induced oral carcinogenesis: a histopathological and biochemical approach. Pathol. Oncol. Res. POR. 2018;24:19–29. doi: 10.1007/s12253-017-0205-7.
    1. Chen YK, Hsue SS, Lin LM. Correlation between inducible nitric oxide synthase and p53 expression for DMBA-induced hamster buccal-pouch carcinomas. Oral. Dis. 2003;9:227–234. doi: 10.1034/j.1601-0825.2003.02878.x.
    1. Li Q, et al. Mouse tumor-bearing models as preclinical study platforms for oral squamous cell carcinoma. Front Oncol. 2020;10:212. doi: 10.3389/fonc.2020.00212.
    1. Ozkok A, Edelstein CL. Pathophysiology of cisplatin-induced acute kidney injury. BioMed. Res. Int. 2014;2014:967826. doi: 10.1155/2014/967826.
    1. Haya-Fernandez MC, Bagan JV, Murillo-Cortes J, Poveda-Roda R, Calabuig C. The prevalence of oral leukoplakia in 138 patients with oral squamous cell carcinoma. Oral. Dis. 2004;10:346–348. doi: 10.1111/j.1601-0825.2004.01031.x.
    1. Mehanna HM, Rattay T, Smith J, McConkey CC. Treatment and follow-up of oral dysplasia - a systematic review and meta-analysis. Head. Neck. 2009;31:1600–1609. doi: 10.1002/hed.21131.
    1. Silverman S, Jr., Gorsky M, Lozada F. Oral leukoplakia and malignant transformation. A follow-up study of 257 patients. Cancer. 1984;53:563–568. doi: 10.1002/1097-0142(19840201)53:3<563::AID-CNCR2820530332>;2-F.
    1. Lorini, L. et al. Overview of oral potentially malignant disorders: from risk factors to specific therapies. Cancers13, 3696 (2021).
    1. Eisen DB, et al. Guidelines of care for the management of actinic keratosis. J. Am. Acad. Dermatol. 2021;85:e209–e233. doi: 10.1016/j.jaad.2021.02.082.
    1. Yardimci G, Kutlubay Z, Engin B, Tuzun Y. Precancerous lesions of oral mucosa. World J. Clin. cases. 2014;2:866–872. doi: 10.12998/wjcc.v2.i12.866.
    1. Lingen MW, DiPietro LA, Solt DB, Bouck NP, Polverini PJ. The angiogenic switch in hamster buccal pouch keratinocytes is dependent on TGFbeta-1 and is unaffected by ras activation. Carcinogenesis. 1997;18:329–338. doi: 10.1093/carcin/18.2.329.
    1. Woolgar JA, Rogers SN, Lowe D, Brown JS, Vaughan ED. Cervical lymph node metastasis in oral cancer: the importance of even microscopic extracapsular spread. Oral. Oncol. 2003;39:130–137. doi: 10.1016/S1368-8375(02)00030-1.
    1. Tran L, et al. Cisplatin alters antitumor immunity and synergizes with PD-1/PD-L1 inhibition in head and neck squamous cell carcinoma. Cancer Immunol. Res. 2017;5:1141–1151. doi: 10.1158/2326-6066.CIR-17-0235.
    1. Tomayko MM, Reynolds CP. Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother. Pharm. 1989;24:148–154. doi: 10.1007/BF00300234.
    1. Jung SH, Lee T, Kim K, George SL. Admissible two-stage designs for phase II cancer clinical trials. Stat. Med. 2004;23:561–569. doi: 10.1002/sim.1600.

Source: PubMed

3
Abonnere