Medical Cannabis in Pediatric Oncology: Friend or Foe?
Abstract
:1. Background
1.1. Pediatric Cancer
1.2. Standard Treatment of Pediatric Cancer
1.3. The Endocannabinoid System and the Developing Brain
1.4. The Endocannabinoid System and Early Brain Development
1.5. Anti-Cancer Mechanisms of Phytocannabinoids
2. Potential Uses of Cannabis in Pediatric Oncology
2.1. Currently Available Cannabis-Based Drugs
2.2. Use of Cannabis in Chemotherapy-Induced Nausea and Vomiting
2.3. Effect of Cannabis-Based Drugs on Quality of Life
2.4. Cannabis’ Anti-Cancer Potential: Pediatric Brain Tumors
2.5. Cannabis’ Anti-Cancer Potential: Pediatric Leukemia
2.6. Cannabis’ Anti-Cancer Potential: Other Pediatric Tumors
3. Concerns about Use of Cannabis in a Pediatric Population
3.1. Early Cannabis Exposure and Brain Development
3.2. Lack of Clinical Studies
3.3. Potential for Addiction
3.4. Physician Perspectives on Pediatric Cannabis Use
3.5. Reports of Adverse Events with Pediatric Cannabis Use
3.6. Regulatory and Legal Issues with Cannabis
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Cancer in Children and Adolescents. Available online: https://www.cancer.gov/types/childhood-cancers/child-adolescent-cancers-fact-sheet (accessed on 6 November 2021).
- Childhood Cancer Statistics. Available online: http://www.cancer.ca/en/cancer-information/cancer-101/childhood-cancer-statistics/?region=on (accessed on 5 November 2021).
- Landier, W.; Armenian, S.H.; Meadows, A.T.; Bhatia, S. Chapter 48—Late Effects of Childhood Cancer and its Treatment. In Principles and Practice of Pediatric Oncology, 7th ed.; Pizzo, P.A., Poplack, D.G., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2016. [Google Scholar]
- Brenner, D.R.; Weir, H.K.; Demers, A.A.; Ellison, L.F.; Louzado, C.; Shaw, A.; Turner, D.; Woods, R.R.; Smith, L.M.; the Canadian Cancer Statistics Advisory Committee. Projected estimates of cancer in Canada in 2020. Can. Med. Assoc. J. (CMAJ) 2020, 192, E199–E205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- American Cancer Society. Cancer Facts & Figures 2020. Available online: https://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2020.html (accessed on 5 November 2021).
- Sheurer, M.E.; Lupo, P.J.; Bondy, M.L. Chapter 1—Epidemiology of Childhood Cancer. In Principles and Practice of Pediatric Oncology, 7th ed.; Pizzo, P.A., Poplack, D.G., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2016. [Google Scholar]
- Denny, C.; Wechsler, D.S.; Sakamoto, K.M. Chapter 4—The Biology of Childhood Cancer. In Principles and Practice of Pediatric Oncology, 7th ed.; Pizzo, P.A., Poplack, D.G., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2016. [Google Scholar]
- Downing, J.R.; Wilson, R.K.; Zhang, J.; Mardis, E.R.; Pui, C.-H.; Ding, L.; Ley, T.J.; Evans, W.E. The Pediatric Cancer Genome Project. Nat. Genet. 2012, 44, 619–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gröbner, S.N.; Worst, B.C.; Weischenfeldt, J.; Buchhalter, I.; Kleinheinz, K.; Rudneva, V.A.; Johann, P.D.; Balasubramanian, G.P.; Segura-Wang, M.; Brabetz, S.; et al. The landscape of genomic alterations across childhood cancers. Nature 2018, 555, 321–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huether, R.; Dong, L.; Chen, X.; Wu, G.; Parker, M.; Wei, L.; Ma, J.; Edmonson, M.N.; Hedlund, E.K.; Rusch, M.C.; et al. The landscape of somatic mutations in epigenetic regulators across 1,000 paediatric cancer genomes. Nat. Commun. 2014, 5, 3630. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Walsh, M.F.; Wu, G.; Edmonson, M.N.; Gruber, T.A.; Easton, J.; Hedges, D.; Ma, X.; Zhou, X.; Yergeau, D.A.; et al. Germline Mutations in Predisposition Genes in Pediatric Cancer. N. Engl. J. Med. 2015, 373, 2336–2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLeod, H.L.; Relling, M.V.; Crom, W.R.; Silverstein, K.; Groom, S.; Rodman, J.H.; Rivera, G.K.; Crist, W.M.; Evans, W.E. Disposition of antineoplastic agents in the very young child. Br. J. Cancer Suppl. 1992, 18, S23–S29. [Google Scholar] [PubMed]
- Adamson, P.C.; Blaney, S.M.; Bagatell, R.; Skolnik, J.M.; Balis, F.M. Chapter 10—General Principles of Chemotherapy. In Principles and Practice of Pediatric Oncology, 7th ed.; Pizzo, P.A., Poplack, D.G., Eds.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2016; pp. 241–315. [Google Scholar]
- Panosyan, E.H.; Ikeda, A.K.; Chang, V.Y.; Laks, D.R.; Reeb, C.L.; Bowles, L.V.; Lasky, R.J.L.; Moore, T.B. High-dose chemotherapy with autologous hematopoietic stem-cell rescue for pediatric brain tumor patients: A single institution experience from UCLA. J. Transplant. 2011, 2011, 740673. [Google Scholar] [CrossRef] [PubMed]
- Shih, C.S.; Hale, G.A.; Gronewold, L.; Tong, X.; Laningham, F.H.; Gilger, E.A.; Srivastava, D.K.; Kun, L.E.; Gajjar, A.; Fouladi, M. High-dose chemotherapy with autologous stem cell rescue for children with recurrent malignant brain tumors. Cancer 2008, 112, 1345–1353. [Google Scholar] [CrossRef] [PubMed]
- Brown, R.J.; Rahim, H.; Wong, K.E.; Cooper, R.M.; Marachelian, A.; Butturini, A.; Dhall, G.; Finlay, J.L. Infectious complications in the first year following autologous hematopoietic progenitor cell rescue for children with brain tumors. Pediatric Blood Cancer 2013, 60, 2012–2017. [Google Scholar] [CrossRef] [PubMed]
- Sands, S.A.; Oberg, J.A.; Gardner, S.L.; Whiteley, J.A.; Glade-Bender, J.L.; Finlay, J.L. Neuropsychological functioning of children treated with intensive chemotherapy followed by myeloablative consolidation chemotherapy and autologous hematopoietic cell rescue for newly diagnosed CNS tumors: An analysis of the Head Start II survivors. Pediatric Blood Cancer 2010, 54, 429–436. [Google Scholar] [CrossRef] [PubMed]
- Ward, E.; DeSantis, C.; Robbins, A.; Kohler, B.; Jemal, A. Childhood and adolescent cancer statistics, 2014. CA A Cancer J. Clin. 2014, 64, 83–103. [Google Scholar] [CrossRef] [PubMed]
- Green, D.M.; Kun, L.E.; Matthay, K.K.; Meadows, A.T.; Meyer, W.H.; Meyers, P.A.; Spunt, S.L.; Robison, L.L.; Hudson, M.M. Relevance of historical therapeutic approaches to the contemporary treatment of pediatric solid tumors. Pediatric Blood Cancer 2013, 60, 1083–1094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hudson, M.M.; Neglia, J.P.; Woods, W.G.; Sandlund, J.T.; Pui, C.H.; Kun, L.E.; Robison, L.L.; Green, D.M. Lessons from the past: Opportunities to improve childhood cancer survivor care through outcomes investigations of historical therapeutic approaches for pediatric hematological malignancies. Pediatric Blood Cancer 2012, 58, 334–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robison, L.L.; Hudson, M.M. Survivors of childhood and adolescent cancer: Life-long risks and responsibilities. Nat. Rev. Cancer 2014, 14, 61–70. [Google Scholar] [CrossRef] [PubMed]
- Kovalchuk, A.; Kolb, B. Chemo brain: From discerning mechanisms to lifting the brain fog—An aging connection. Cell Cycle (Georget. Tex.) 2017, 16, 1345–1349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maida, V.; Daeninck, P.J. A user’s guide to cannabinoid therapies in oncology. Curr. Oncol. 2016, 23, 398–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Marzo, V. New approaches and challenges to targeting the endocannabinoid system. Nat. Rev. Drug Discov. 2018, 17, 623–639. [Google Scholar] [CrossRef] [PubMed]
- Young, A.C.; Lolait, S.J.; Brownstein, M.J.; Matsuda, L.A.; Bonner, T.I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990, 346, 561–564. [Google Scholar]
- Munro, S.; Abu-Shaar, M.; Thomas, K.L. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef] [PubMed]
- Pisanti, S.; Malfitano, A.M.; Ciaglia, E.; Lamberti, A.; Ranieri, R.; Cuomo, G.; Abate, M.; Faggiana, G.; Proto, M.C.; Fiore, D.; et al. Cannabidiol: State of the art and new challenges for therapeutic applications. Pharmacol. Ther. 2017, 175, 133–150. [Google Scholar] [CrossRef] [PubMed]
- Muller, C.; Morales, P.; Reggio, P.H. Cannabinoid Ligands Targeting TRP Channels. Front. Mol. Neurosci. 2019, 11, 487. [Google Scholar] [CrossRef] [PubMed]
- Kovalchuk, O.; Kovalchuk, I. Cannabinoids as anticancer therapeutic agents. Cell Cycle (Georget. Tex.) 2020, 19, 961–989. [Google Scholar] [CrossRef] [PubMed]
- Gaoni, Y.; Mechoulam, R. Isolation, structure, and partial synthesis of an active constituent of hashish. J. Am. Chem. Soc. 1964, 86, 1646–1647. [Google Scholar] [CrossRef]
- Mechoulam, R.; Shvo, Y. Hashish—I: The structure of Cannabidiol. Tetrahedron 1963, 19, 2073–2078. [Google Scholar] [CrossRef]
- Alves, P.; Amaral, C.; Teixeira, N.; Correia-da-Silva, G. Cannabis sativa: Much more beyond Delta(9)-tetrahydrocannabinol. Pharmacol. Res. 2020, 157, 104822. [Google Scholar] [CrossRef] [PubMed]
- Tomko, A.M.; Whynot, E.G.; Ellis, L.D.; Dupre, D.J. Anti-Cancer Potential of Cannabinoids, Terpenes, and Flavonoids Present in Cannabis. Cancers 2020, 12, 1985. [Google Scholar] [CrossRef] [PubMed]
- Fride, E. Multiple Roles for the Endocannabinoid System During the Earliest Stages of Life: Pre- and Postnatal Development. J. Neuroendocrinol. 2008, 20, 75–81. [Google Scholar] [CrossRef] [PubMed]
- Fernández-Ruiz, J.J.; Berrendero, F.; Hernández, M.L.; Romero, J.; Ramos, J.A. Role of endocannabinoids in brain development. Life Sci. 1999, 65, 725–736. [Google Scholar] [CrossRef]
- Mato, S.; Del Olmo, E.; Pazos, A. Ontogenetic development of cannabinoid receptor expression and signal transduction functionality in the human brain. Eur. J. Neurosci. 2003, 17, 1747–1754. [Google Scholar] [CrossRef]
- Vitalis, T.; Lainé, J.; Simon, A.; Roland, A.; Leterrier, C.; Lenkei, Z. The type 1 cannabinoid receptor is highly expressed in embryonic cortical projection neurons and negatively regulates neurite growth in vitro. Eur. J. Neurosci. 2008, 28, 1705–1718. [Google Scholar] [CrossRef]
- Harkany, T.; Mackie, K.; Doherty, P. Wiring and firing neuronal networks: Endocannabinoids take center stage. Curr. Opin. Neurobiol. 2008, 18, 338–345. [Google Scholar] [CrossRef] [Green Version]
- Fride, E. The endocannabinoid-CB1 receptor system in pre- and postnatal life. Eur. J. Pharmacol. 2004, 500, 289–297. [Google Scholar] [CrossRef]
- Munson, A.E.; Harris, L.S.; Friedman, M.A.; Dewey, W.L.; Carchman, R.A. Antineoplastic Activity of Cannabinoids. JNCI J. Natl. Cancer Inst. 1975, 55, 597–602. [Google Scholar] [CrossRef] [PubMed]
- Sledzinski, P.; Zeyland, J.; Slomski, R.; Nowak, A. The current state and future perspectives of cannabinoids in cancer biology. Cancer Med. 2018, 7, 765–775. [Google Scholar] [CrossRef] [PubMed]
- Baram, L.; Peled, E.; Berman, P.; Yellin, B.; Besser, E.; Benami, M.; Louria-Hayon, I.; Lewitus, G.M.; Meiri, D. The heterogeneity and complexity of Cannabis extracts as antitumor agents. Oncotarget 2019, 10, 4091–4106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scott, K.A.; Dalgleish, A.G.; Liu, W.M. Anticancer effects of phytocannabinoids used with chemotherapy in leukaemia cells can be improved by altering the sequence of their administration. Int. J. Oncol. 2017, 51, 369–377. [Google Scholar] [CrossRef] [Green Version]
- McPartland, J.M.; Russo, E.B. Cannabis and Cannabis Extracts: Greater Than the Sum of Their Parts? J. Cannabis Ther. 2001, 1, 103–132. [Google Scholar] [CrossRef]
- Pisanti, S.; Picardi, P.; D’Alessandro, A.; Laezza, C.; Bifulco, M. The endocannabinoid signaling system in cancer. Trends Pharmacol. Sci. 2013, 34, 273–282. [Google Scholar] [CrossRef]
- Velasco, G.; Hernández-Tiedra, S.; Dávila, D.; Lorente, M. The use of cannabinoids as anticancer agents. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2016, 64, 259–266. [Google Scholar] [CrossRef] [Green Version]
- Velasco, G.; Sanchez, C.; Guzman, M. Anticancer mechanisms of cannabinoids. Curr. Oncol. (Tor. Ont.) 2016, 23, S23–S32. [Google Scholar] [CrossRef] [Green Version]
- Pyszniak, M.; Tabarkiewicz, J.; Luszczki, J.J. Endocannabinoid system as a regulator of tumor cell malignancy—Biological pathways and clinical significance. Oncotargets Ther. 2016, 9, 4323–4336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nikan, M.; Nabavi, S.M.; Manayi, A. Ligands for cannabinoid receptors, promising anticancer agents. Life Sci. 2016, 146, 124–130. [Google Scholar] [CrossRef] [PubMed]
- Likar, R.; Nahler, G. The use of cannabis in supportive care and treatment of brain tumor. Neuro-Oncol. Pract. 2017, 4, 151–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Calvaruso, G.; Pellerito, O.; Notaro, A.; Giuliano, M. Cannabinoid-associated cell death mechanisms in tumor models (Review). Int. J. Oncol. 2012, 41, 407–413. [Google Scholar] [CrossRef] [Green Version]
- Mukhopadhyay, B.; Schuebel, K.; Mukhopadhyay, P.; Cinar, R.; Godlewski, G.; Xiong, K.; Mackie, K.; Lizak, M.; Yuan, Q.; Goldman, D.; et al. Cannabinoid receptor 1 promotes hepatocellular carcinoma initiation and progression through multiple mechanisms. Hepatology 2014, 61, 1615–1626. [Google Scholar] [CrossRef]
- Marshall, A.D.; Lagutina, I.; Grosveld, G.C. PAX3-FOXO1 Induces Cannabinoid Receptor 1 to Enhance Cell Invasion and Metastasis. Cancer Res. 2011, 71, 7471–7480. [Google Scholar] [CrossRef] [Green Version]
- Hart, S.; Fischer, O.M.; Ullrich, A. Cannabinoids induce cancer cell proliferation via tumor necrosis factor alpha-converting enzyme (TACE/ADAM17)-mediated transactivation of the epidermal growth factor receptor. Cancer Res. 2004, 64, 1943–1950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freund, P.; Porpaczy, E.A.; Le, T.; Gruber, M.; Pausz, C.; Staber, P.; Jager, U.; Vanura, K. Cannabinoid Receptors Are Overexpressed in CLL but of Limited Potential for Therapeutic Exploitation. PLoS ONE 2016, 11, e0156693. [Google Scholar] [CrossRef] [Green Version]
- Prevarskaya, N.; Zhang, L.; Barritt, G. TRP channels in cancer. Biochim. Biophys. Acta-Mol. Basis Dis. 2007, 1772, 937–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gunasekera, B.; Davies, C.; Martin-Santos, R.; Bhatthacharyya, S. The Yin and Yang of Cannabis-a systematic review of human neuroimaging evidence of the differential effects of delta-9-tetrahydrocannabinol and cannabidiol. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2020, 6, 636–645. [Google Scholar]
- Wong, S.S.; Wilens, T.E. Medical Cannabinoids in Children and Adolescents: A Systematic Review. Pediatrics 2017, 140, e20171818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ananth, P.; Reed-Weston, A.; Wolfe, J. Medical marijuana in pediatric oncology: A review of the evidence and implications for practice. Pediatric Blood Cancer 2017, 65, e26826. [Google Scholar] [CrossRef] [PubMed]
- Mitelpunkt, A.; Kramer, U.; Kedem, M.H.; Fink, E.Z.; Orbach, R.; Chernuha, V.; Fattal-Valevski, A.; Deutsch, L.; Heffetz, D.; Sacks, H. The safety, tolerability, and effectiveness of PTL-101, an oral cannabidiol formulation, in pediatric intractable epilepsy: A phase II, open-label, single-center study. Epilepsy Behav. 2019, 98, 233–237. [Google Scholar] [CrossRef] [PubMed]
- Fairhurst, C.; Kumar, R.; Checketts, D.; Tayo, B.; Turner, S. Efficacy and safety of nabiximols cannabinoid medicine for paediatric spasticity in cerebral palsy or traumatic brain injury: A randomized controlled trial. Dev. Med. Child Neurol. 2020, 62, 1031–1039. [Google Scholar] [CrossRef] [PubMed]
- Wheless, J.W.; Dlugos, D.; Miller, I.; Oh, D.A.; Parikh, N.; Phillips, S.; Renfroe, J.B.; Roberts, C.M.; Saeed, I.; Sparagana, S.P.; et al. Pharmacokinetics and Tolerability of Multiple Doses of Pharmaceutical-Grade Synthetic Cannabidiol in Pediatric Patients with Treatment-Resistant Epilepsy. CNS Drugs 2019, 33, 593–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Devinsky, O.; Patel, A.D.; Thiele, E.A.; Wong, M.H.; Appleton, R.; Harden, C.L.; Greenwood, S.; Morrison, G.; Sommerville, K. Randomized, dose-ranging safety trial of cannabidiol in Dravet syndrome. Neurology 2018, 90, e1204–e1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Devinsky, O.; Cross, J.H.; Laux, L.; Marsh, E.; Miller, I.; Nabbout, R.; Scheffer, I.E.; Thiele, E.A.; Wright, S. Trial of Cannabidiol for Drug-Resistant Seizures in the Dravet Syndrome. N. Engl. J. Med. 2017, 376, 2011–2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, I.; Scheffer, I.E.; Gunning, B.; Sanchez-Carpintero, R.; Gil-Nagel, A.; Perry, M.S.; Saneto, R.P.; Checketts, D.; Dunayevich, E.; Knappertz, V. Dose-Ranging Effect of Adjunctive Oral Cannabidiol vs Placebo on Convulsive Seizure Frequency in Dravet Syndrome: A Randomized Clinical Trial. JAMA Neurol. 2020, 77, 613–621. [Google Scholar] [CrossRef]
- Devinsky, O.; Patel, A.D.; Cross, J.H.; Villanueva, V.; Wirrell, E.C.; Privitera, M.; Greenwood, S.M.; Roberts, C.; Checketts, D.; Van Landingham, K.E.; et al. Effect of Cannabidiol on Drop Seizures in the Lennox-Gastaut Syndrome. N. Engl. J. Med. 2018, 378, 1888–1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thiele, E.A.; Marsh, E.D.; French, J.A.; Mazurkiewicz-Beldzinska, M.; Benbadis, S.R.; Joshi, C.; Lyons, P.D.; Taylor, A.; Roberts, C.; Sommerville, K. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2018, 391, 1085–1096. [Google Scholar] [CrossRef]
- Thiele, E.; Marsh, E.; Mazurkiewicz-Beldzinska, M.; Halford, J.J.; Gunning, B.; Devinsky, O.; Checketts, D.; Roberts, C. Cannabidiol in patients with Lennox-Gastaut syndrome: Interim analysis of an open-label extension study. Epilepsia 2019, 60, 419–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thiele, E.A.; Bebin, E.M.; Bhathal, H.; Jansen, F.E.; Kotulska, K.; Lawson, J.A.; O’Callaghan, F.J.; Wong, M.; Sahebkar, F.; Checketts, D. Add-On Cannabidiol Treatment for Drug-Resistant Seizures in Tuberous Sclerosis Complex: A Placebo-Controlled Randomized Clinical Trial. JAMA Neurol. 2020, 78, 285–292. [Google Scholar] [CrossRef] [PubMed]
- Aran, A.; Harel, M.; Cassuto, H.; Polyansky, L.; Schnapp, A.; Wattad, N.; Shmueli, D.; Golan, D.; Castellanos, F.X. Cannabinoid treatment for autism: A proof-of-concept randomized trial. Mol. Autism 2021, 12, 6. [Google Scholar] [CrossRef] [PubMed]
- Devinsky, O.; Marsh, E.; Friedman, D.; Thiele, E.; Laux, L.; Sullivan, J.; Miller, I.; Flamini, R.; Wilfong, A.; Filloux, F.; et al. Cannabidiol in patients with treatment-resistant epilepsy: An open-label interventional trial. Lancet Neurol. 2016, 15, 270–278. [Google Scholar] [CrossRef]
- Devinsky, O.; Nabbout, R.; Miller, I.; Laux, L.; Zolnowska, M.; Wright, S.; Roberts, C. Long-term cannabidiol treatment in patients with Dravet syndrome: An open-label extension trial. Epilepsia 2019, 60, 294–302. [Google Scholar] [CrossRef]
- Porter, B.E.; Jacobson, C. Report of a parent survey of cannabidiol-enriched cannabis use in pediatric treatment-resistant epilepsy. Epilepsy Behav. 2013, 29, 574–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silvestro, S.; Mammana, S.; Cavalli, E.; Bramanti, P.; Mazzon, E. Use of cannabidiol in the treatment of epilepsy: Efficacy and security in clinical trials. Molecules 2019, 24, 1459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galan, F.N.; Miller, I. Cannabinoids for the Treatment of Epilepsy: A Review. Curr. Treat. Options Neurol. 2020, 22, 1–14. [Google Scholar] [CrossRef]
- Gaston, T.E.; Szaflarski, J.P. Cannabis for the Treatment of Epilepsy: An Update. Curr. Neurol. Neurosci. Rep. 2018, 18, 1–9. [Google Scholar] [CrossRef]
- Geffrey, A.L.; Pollack, S.F.; Bruno, P.L.; Thiele, E.A. Drug-drug interaction between clobazam and cannabidiol in children with refractory epilepsy. Epilepsia (Copenhagen) 2015, 56, 1246–1251. [Google Scholar] [CrossRef]
- Kuhlen, M.; Hoell, J.I.; Gagnon, G.; Balzer, S.; Oommen, P.T.; Borkhardt, A.; Janßen, G. Effective treatment of spasticity using dronabinol in pediatric palliative care. Eur. J. Paediatr. Neurol. 2016, 20, 898–903. [Google Scholar] [CrossRef] [PubMed]
- Yeung, M.; Wroot, H.; Charnock, C.; Forbes, C.; Lafay-Cousin, L.; Schulte, F. Cannabis use in pediatric cancer patients: What are they reading? A review of the online literature. Support. Care Cancer Off. J. Multinatl. Assoc. Support. Care Cancer 2020, 28, 3503–3515. [Google Scholar] [CrossRef] [PubMed]
- Andradas, C.; Truong, A.; Byrne, J.; Endersby, R. The Role of Cannabinoids as Anticancer Agents in Pediatric Oncology. Cancers 2021, 13, 157. [Google Scholar] [CrossRef] [PubMed]
- Borgelt, L.M.; Franson, K.L.; Nussbaum, A.M.; Wang, G.S. The pharmacologic and clinical effects of medical cannabis. Pharmacotherapy 2013, 33, 195–209. [Google Scholar] [CrossRef] [PubMed]
- Pertwee, R.G. Emerging strategies for exploiting cannabinoid receptor agonists as medicines. Br. J. Pharmacol. 2009, 156, 397–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schuman, A.J. Cannabidiol use in children. Contemp. Pediatrics 2019, 36, 40–44. [Google Scholar]
- Phillips, R.S.; Friend, A.J.; Gibson, F.; Houghton, E.; Gopaul, S.; Craig, J.V.; Pizer, B. Antiemetic medication for prevention and treatment of chemotherapy-induced nausea and vomiting in childhood. Cochrane Database Syst. Rev. 2016, 2, CD007786. [Google Scholar] [CrossRef]
- Dalzell, A.M.; Bartlett, H.; Lilleyman, J.S. Nabilone: An alternative antiemetic for cancer chemotherapy. Arch. Dis. Child. 1986, 61, 502–505. [Google Scholar] [CrossRef] [PubMed]
- Chan, H.S.L.; Correia, J.A.; MacLeod, S.M. Nabilone Versus Prochlorperazine for Control of Cancer Chemotherapy-Induced Emesis in Children: A Double-Blind, Crossover Trial. Pediatrics 1987, 79, 946–952. [Google Scholar] [PubMed]
- Ekert, H.; Waters, K.D.; Jurk, I.H.; Mobilia, J.; Loughnan, P. Amelioration of cancer chemotherapy-induced nausea and vomiting by delta-9-tetrahydro-cannabinol. Med. J. Aust. 1979, 2, 657–659. [Google Scholar] [CrossRef]
- Abrahamov, A.; Abrahamov, A.; Mechoulam, R. An efficient new cannabinoid antiemetic in pediatric oncology. Life Sci. 1995, 56, 2097–2102. [Google Scholar] [CrossRef]
- Ofir, R.; Bar-Sela, G.; Weyl Ben-Arush, M.; Postovsky, S. Medical marijuana use for pediatric oncology patients: Single institution experience. Pediatric Hematol. Oncol. 2019, 36, 255–266. [Google Scholar] [CrossRef] [PubMed]
- Sredni, S.T.; Huang, C.-C.; Suzuki, M.; Pundy, T.; Chou, P.; Tomita, T. Spontaneous involution of pediatric low-grade gliomas: High expression of cannabinoid receptor 1 (CNR1) at the time of diagnosis may indicate involvement of the endocannabinoid system. Child’s Nerv. Syst. 2016, 32, 2061–2067. [Google Scholar] [CrossRef] [PubMed]
- Ellert-Miklaszewska, A.; Grajkowska, W.; Gabrusiewicz, K.; Kaminska, B.; Konarska, L. Distinctive pattern of cannabinoid receptor type II (CB2) expression in adult and pediatric brain tumors. Brain Res. 2006, 1137, 161–169. [Google Scholar] [CrossRef] [PubMed]
- Fisher, T.; Golan, H.; Schiby, G.; PriChen, S.; Smoum, R.; Moshe, I.; Peshes-Yaloz, N.; Castiel, A.; Waldman, D.; Gallily, R.; et al. In vitro and in vivo efficacy of non-psychoactive cannabidiol in neuroblastoma. Curr. Oncol. 2016, 23, S15–S22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alharris, E.; Singh, N.P.; Nagarkatti, P.S.; Nagarkatti, M. Role of miRNA in the regulation of cannabidiol-mediated apoptosis in neuroblastoma cells. Oncotarget 2019, 10, 45–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caballero, F.J.; Soler-Torronteras, R.; Lara-Chica, M.; Garcia, V.; Fiebich, B.L.; Munoz, E.; Calzado, M.A. AM404 inhibits NFAT and NF-kappa B signaling pathways and impairs migration and invasiveness of neuroblastoma cells. Eur. J. Pharmacol. 2015, 746, 221–232. [Google Scholar] [CrossRef] [PubMed]
- McKallip, R.J.; Jia, W.; Schlomer, J.; Warren, J.W.; Nagarkatti, P.S.; Nagarkatti, M. Cannabidiol-induced apoptosis in human leukemia cells: A novel role of cannabidiol in the regulation of p22phox and Nox4 expression. Mol. Pharmacol. 2006, 70, 897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lombard, C.; Nagarkatti, M.; Nagarkatti, P.S. Targeting cannabinoid receptors to treat leukemia: Role of cross-talk between extrinsic and intrinsic pathways in Delta(9)-tetrahydrocannabinol (THC)-induced apoptosis of Jurkat cells. Leuk. Res. 2005, 29, 915–922. [Google Scholar] [CrossRef]
- Jia, W.; Hegde, V.L.; Singh, N.P.; Sisco, D.; Grant, S.; Nagarkatti, M.; Nagarkatti, P.S. Delta 9-tetrahydrocannabinol-induced apoptosis in Jurkat leukemia T cells is regulated by translocation of bad to mitochondria. Mol. Cancer Res. 2006, 4, 549–562. [Google Scholar] [CrossRef] [Green Version]
- Herrera, B.; Carracedo, A.; Diez-Zaera, M.; del Pulgar, T.G.; Guzman, M.; Velasco, G. The CB2 cannabinoid receptor signals apoptosis via ceramide-dependent activation of the mitochondrial intrinsic pathway. Exp. Cell Res. 2006, 312, 2121–2131. [Google Scholar] [CrossRef] [PubMed]
- Soto-Mercado, V.; Mendivil-Perez, M.; Jimenez-Del-Rio, M.; Fox, J.E.; Velez-Pardo, C. Cannabinoid CP55940 selectively induces apoptosis in Jurkat cells and in ex vivo T-cell acute lymphoblastic leukemia through H2O2 signaling mechanism. Leuk. Res. 2020, 95, 12. [Google Scholar] [CrossRef] [PubMed]
- Olivas-Aguirre, M.; Torres-Lopez, L.; Valle-Reyes, J.S.; Hernandez-Cruz, A.; Pottosin, I.; Dobrovinskaya, O. Cannabidiol directly targets mitochondria and disturbs calcium homeostasis in acute lymphoblastic leukemia. Cell Death Dis. 2019, 10, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, Y.; Bali, C. Cannabis Extract Treatment for Terminal Acute Lymphoblastic Leukemia with a Philadelphia Chromosome Mutation. Case Rep. Oncol. 2013, 6, 585–592. [Google Scholar] [CrossRef] [PubMed]
- Foroughi, M.; Hendson, G.; Sargent, M.A.; Steinbok, P. Spontaneous regression of septum pellucidum/forniceal pilocytic astrocytomas—possible role of Cannabis inhalation. Child’s Nerv. Syst. 2011, 27, 671–679. [Google Scholar] [CrossRef] [PubMed]
- Oesch, S.; Walter, D.; Wachtel, M.; Pretre, K.; Salazar, M.; Guzmán, M.; Velasco, G.; Schäfer, B.W. Cannabinoid receptor 1 is a potential drug target for treatment of translocation-positive rhabdomyosarcoma. Mol. Cancer Ther. 2009, 8, 1838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernard, C.; Milh, M.; Morozov, Y.M.; Ben-Ari, Y.; Freund, T.F.; Gozlan, H.; Miledi, R. Altering Cannabinoid Signaling during Development Disrupts Neuronal Activity. Proc. Natl. Acad. Sci. USA 2005, 102, 9388–9393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meyer, H.C.; Lee, F.S.; Gee, D.G. The Role of the Endocannabinoid System and Genetic Variation in Adolescent Brain Development. Neuropsychopharmacology 2018, 43, 21–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camchong, J.; Lim, K.O.; Kumra, S. Adverse Effects of Cannabis on Adolescent Brain Development: A Longitudinal Study. Cereb. Cortex (New York N.Y. 1991) 2017, 27, 1922–1930. [Google Scholar] [CrossRef] [Green Version]
- Ananth, P.; Ma, C.; Al-Sayegh, H.; Kroon, L.; Klein, V.; Wharton, C.; Hallez, E.; Braun, I.; Michelson, K.; Rosenberg, A.R.; et al. Provider Perspectives on Use of Medical Marijuana in Children with Cancer. Pediatrics 2018, 141, e20170559. [Google Scholar] [CrossRef] [Green Version]
- Golan, H.; Fisher, T.; Toren, A. The Role of Cannabinoids in the Treatment of Children with Cancer. Isr. Med. Assoc. J. 2017, 19, 89–94. [Google Scholar] [PubMed]
- Schonhofen, P.; Bristot, I.J.; Crippa, J.A.; Hallak, J.E.C.; Zuardi, A.W.; Parsons, R.B.; Klamt, F. Cannabinoid-Based Therapies and Brain Development: Potential Harmful Effect of Early Modulation of the Endocannabinoid System. CNS Drugs 2018, 32, 697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Government of Canada. Cannabis Market Data: Overview. Available online: https://www.canada.ca/en/health-canada/services/drugs-medication/cannabis/research-data/market.html (accessed on 20 July 2021).
Identifier | Title | Age of Patients (Years) | Disease | Study Type | Cannabinoid Drug Used | Concomitant Treatment? | Results | Side Effects | Ref |
---|---|---|---|---|---|---|---|---|---|
NCT02987114 | A Phase II, Open-label, Single-center Clinical Study to Evaluate the Safety, Tolerability, and Efficacy of Oral Administration of PTL101 | 2–15 | TRE | Interventional, single group assessment, open label | CBD capsules (PTL101), twice daily, 25 mg/kg/day up to 450 mg/kg/day | - | 81.9% ± 24.6% reduction baseline median seizures, 73.4 ± 24.6% reduction monthly seizure frequency | Sleep disturbance/insomnia 25%, somnolence 18.8%, increased seizure frequency 18.8%, restlessness 18.8%, no SAE | [60] |
NCT01898520 | The Efficacy, Safety and Tolerability of Sativex as an Adjunctive Treatment to Existing Anti-spasticity Medications in Children Aged 8 to 18 Years With Spasticity Due to Cerebral Palsy or Traumatic Central Nervous System Injury Who Have Not Responded Adequately to Their Existing Anti-spasticity Medications: a Parallel Group Randomised, Double-blind, Placebo-controlled Study Followed by a 24-week Open Label | 8–18 | CP or traumatic CNS injury | Interventional, parallel assignment, double-blind, placebo-controlled | Sativex (THC 27 mg/mL: CBD 25 mg/mL), dose of 2.7 mg THC and 2.5 mg CBD, up to 12 doses per day | - | No difference in caregiver-reported spasticity | 39% had treatment-related adverse events, only 23% in placebo-controlled group | [61] |
NCT02324673 | A Phase ½ Study to assess the pharmacokinetics and safety of Multiple Doses of Pharmaceutical Cannabidiol Oral Solution in Pediatric Patients with Treatment-Resistant Seizure Disorders | 1–17 | TRS | Interventional, randomized, parallel assignment, placebo controlled, double blind | CBD at 10, 20, or 40 mg/kg/day | - | CBD levels variable across patients, but overall well tolerated. | Somnolence 21.3%, anemia 18%, diarrhea 16.4%; 3 (1.6%) SAEs related to study drug: skin rash. | [62] |
NCT02091206 | A Double Blind, Placebo-controlled, Two-part Study to Investigate the Dose-ranging Safety and Pharmacokinetics, Followed by the Efficacy and Safety of Cannabidiol (GWP42003-P) in Children and Young Adults With Dravet Syndrome | 4–10 | DS | Interventional, randomized, parallel assignment, placebo controlled, double blind | CBD (Epidiolex) at either 5, 10, or 20 mg/kg/day | CBZ, LVA, SPL, TPM, VPA | When taking AED with CBD, CBD did not affect AED levels | Pyrexia, somnolence, decreased appetite, sedation, vomiting, ataxia, and abnormal behavior. Two discontinued for AEs: pyrexia and rash, and elevated transaminases | [63] |
NCT02091375 | A Double Blind, Placebo Controlled Two-part Study to Investigate the Dose-ranging Safety and Pharmacokinetics, Followed by the Efficacy and Safety of Cannabidiol (GWP42003-P) in Children and Young Adults With Dravet Syndrome | 2–18 | DS | Interventional, randomized, parallel assignment, placebo controlled, double blind | Epidiolex 20 mg/kg/day | CBZ, LVA, SPL, TPM, VPA | A total of 43% of patients had 50% reduction in convulsive seizure frequency, frequency of all seizures was significantly reduced, excepting nonconvulsive seizures | Diarrhea, vomiting, fatigue, pyrexia, somnolence, and abnormal liver-function tests, more withdrawals in CBD group; 93% of CBD group experienced AE vs. 75% placebo, SAE for 16.4% CBD patients and 5.1% placebo | [64] |
NCT02224703 | A Randomized, Double-blind, Placebo-controlled Study to Investigate the Efficacy and Safety of Cannabidiol (GWP42003-P) in Children and Young Adults With Dravet Syndrome | 2–18 | DS | Interventional, randomized, parallel assignment, placebo controlled, double blind | CBD (Epidiolex) at either 10 or 20 mg/kg/day | - | Reduction from baseline in convulsive seizures: 48.7% for 20 mg/kg/day, 45.7% for 10 mg/kg/day, 29.8% placebo. Reduction from baseline in total seizure frequency: 47.3% for 20 mg/kg/day, 56.4% for 10 mg/kg/day, 29.7% placebo | A total of 88% experienced adverse events (89.9% 20 mg/kg/day, 87.% 10 mg/kg/day, 89.2% placebo); decreased appetite, diarrhea, somnolence, pyrexia, fatigue; 22.7% SAEs; 51.7% AEs resolved by end of trial for CBD, 60.3% resolved for placebo | [65] |
NCT02224560 | A Randomized, Double-blind, Placebo-controlled Study to Investigate the Efficacy and Safety of Cannabidiol (GWP42003-P; CBD) as Adjunctive Treatment for Seizures Associated With Lennox-Gastaut Syndrome in Children and Adults. | 2–55 | LGS | Interventional, randomized, parallel assignment, placebo controlled, double blind | Epidiolex, either 10 or 20 mg/kg/day | CBZ, LVA, SPL, TPM, VPA | Patients receiving 20 mg/kg/day had 41.9% reduction in drop seizures and those receiving 10 mg/kg/day had 37.2% reduction, placebo had 17.2% reduction | AEs occurred for 94% of 20 mg group, 84% of 10 mg group, and 72% of placebo group; 89% of all AEs mild (somnolence, decreased appetite, pyrexia, vomiting); 26 had SAE in either CBD group, 7 of which were related to CBD | [66] |
NCT02224690 | A Randomized, Double-blind, Placebo-controlled Study to Investigate the Efficacy and Safety of Cannabidiol (GWP42003-P) as Adjunctive Treatment for Seizures Associated with Lennox-Gastaut Syndrome in Children and Adults | 2–55 | LGS | Interventional, randomized, parallel assignment, placebo controlled, double blind | Epidiolex, 20 mg/kg/day | CBZ, LVA, SPL, TPM, VPA | Median reduction in monthly drop seizure frequency from baseline 43.9% in CBD group, 21.8% in placebo. | AEs in 86% of patients in CBD group, 69% in placebo group, most mild/moderate (diarrhea, somnolence, pyrexia, decreased appetite, vomiting). | [67] |
NCT02224573 | An Open-Label Extension Study to Investigate the Safety of Cannabidiol (GWP42003-P; CBD) in Children and Young Adults with Inadequately Controlled Dravet or Lennox-Gastaut Syndrome | Over 2 years and completed NCT02224960, NCT02224560, NCT02224703, NCT02091206, or NCT02091375 | DS, LGS | Interventional, single group assignment, open label | Epidiolex, concentration not specified | CBZ, LVA, SPL, TPM, VPA | Median reduction from baseline in drop seizure frequency from 48–60% over 48 weeks, median reduction in monthly seizure frequency 48–57% over 48 weeks. | A total of 92.1% had adverse events, 32.5% mild, and 43.4% moderate (diarrhea, somnolence, convulsion); 9.6% discontinued due to AEs. | [68] |
NCT02544763 | A Double-blind, Randomized, Placebo-controlled Study to Investigate the Efficacy and Safety of Cannabidiol (GWP42003-P, CBD) as Add-on Therapy in Patients with Tuberous Sclerosis Complex Who Experience Inadequately Controlled Seizures | 1–65 | TCS | Interventional, randomized, parallel assignment, placebo-controlled, double-blind | CBD solution, either 25 or 50 mg/kg/day | CBZ, LVA, VPA, VGB | Percent reduction from baseline in seizures 47.5% for 50 mg/kg/day CBD, 48.56% for 25 mg/kg/day CBD, 26.5% for placebo | Diarrhea and somnolence occurred more for CBD groups than placebo, and more in 50 mg/kg/day group than 25 mg/kg/day group. All patients in 50 mg/kg/day group experienced AEs. SAEs in 14% 50 mg/kg/day, 21% 25 mg/kg/day, 3% placebo | [69] |
NCT02956226 | Cannabinoids for Behavioral Problems in Autism Spectrum Disorder: A Double Blind, Randomized, Placebo-controlled Trial with Crossover | 5–21 | ASD | Interventional, randomized, parallel assignment, placebo-controlled, double-blind | Mix of pure CBD and THC in 20:1 ratio, or whole-plant extract enriched in CBD and THC to 20:1 ratio. Dose maximum 10 mg/kg/day | - | No change in behavioral problems generally, but improvement of disruptive behavior 49% on whole-plant extract vs. placebo. | No SAEs. Common adverse events were somnolence (28% plant extract, 23% pure cannabinoids, 8% placebo) and decreased appetite (25% plant extract, 21% pure cannabinoids, and 15% placebo) | [70] |
Identifier | Title | Patients | Disease | Study Type | Cannabinoid Drug Used | Status |
---|---|---|---|---|---|---|
NCT03467113 | A study to assess the safety and tolerability of ZX008 in children and young adults with DS or LGS currently taking CBD | Children (2–18) | Dravet or Lennox–Gastaut syndromes | Interventional, single group assignment, open label | ZX008, fenfluramine hydrochloride 0.2 or 0.8 mg/day, but patients must also be taking CBD | ANR |
NCT03024827 | Cannabidiol in children with refractory epileptic encephalopathy (CARE-E) | Children (1–10) | Epileptic encephalopathy | Interventional, single group assignment, open label | Cannimed 1:20 (CBD:THC) | ANR |
NCT02523183 | The use of medicinal cannabinoids as adjunctive treatment for medically refractory epilepsy | Children and young adults (0–20) | Medically refractory epilepsy | Observational, cohort, prospective | Medical cannabis | ANR |
NCT02983695 | Cannabinoid Therapy for Pediatric Epilepsy | Children (1–18) | Dravet syndrome | Interventional, single group assignment, open label | TIL-TC150 (contains THC and CBD) | ANR |
NCT02660255 | Safety and Tolerability of Cannabidiol in Subjects with Drug-Resistant Epilepsy | Children and adults (1–60) | Drug-resistant epilepsy | Expanded access | CBD (epidiolex) | AfM |
NCT02397863 | Epidiolex and Drug-Resistant Epilepsy in Children | Children (1–18) | Drug-resistant epilepsy (excluding Dravet Syndrome or Lennox–Gastaut Syndrome) | Expanded access | CBD (epidiolex, 25 mg/kg/day, up to 50 mg/kg/day) | A |
NCT03196934 | Expanded use of cannabidiol oral solution | Children (1–18) | Treatment-resistant seizure disorder | Expanded access | CBD | A |
NCT03676049 | Cannabidiol for drug-resistant pediatric epilepsy (expanded access use) | Children and young adults (5–19) | Drug-resistant epilepsy | Expanded access | CBD, between 2.5 and 7.5 mg/kg/day | A |
NCT02987114 | A study to evaluate the safety, tolerability, and efficacy of oral administration of PTL101 (Cannabidiol) as an adjunctive treatment for pediatric intractable epilepsy | Children (2–15) | Refractory epilepsy | Interventional, single group assignment, open label | Cannabidiol (PTL101), capsules taken twice daily. Capsules are either 50 or 100 mg and taken up to 25 mg/kg/day or up to 450 mg/kg/day | CNP |
NCT02286986 | Cannabidiol (CBD) to 27 patients (aged 2–19 years) with drug-resistant epilepsy | Children and young adults (2–25) | Drug-resistant epilepsy | Interventional, open label, single group assignment | Cannabidiol | CNP |
NCT01898520 | A safety, efficacy, and tolerability study of Sativex for the treatment of spasticity in children aged 8–18 years | Children (8–18) | Patients with CP or traumatic CNS injury with non-progressive spasticity | Interventional, parallel assignment, double-blind, placebo-controlled | THC:CBD 27:25 mg/mL spray (Sativex), given 100 uL per spray up to 12 per day | CNP |
NCT02224573 | GWPCARE5- an open-label extension study of cannabidiol (GWP42003-P) in children and young adults with Dravet or Lennox–Gastaut syndrome | Children and adults (2+) | Lennox–Gastaut syndrome, Dravet syndrome | Interventional, single group assignment, open label | Epidiolex | CNP |
NCT02953548 | Trial of cannabidiol (CBD; GWP42003-P) for infantile spasms | Children (0.083–2) | Infantile spasms and hypsarrhythmia | Interventional, single group assignment, open label | Epidiolex, 40 mg/kg/day | CNP |
NCT02954887 | Phase 3 trial of cannabidiol (CBD; GWP42003-P) for infantile spasms: open-label extension phase (GWPCARE7) | Children (0.083–2) | Infantile spasms and hypsarrhythmia | Interventional, single group assignment, open label | Epidiolex, 40 mg/kg/day | CNP |
NCT02073474 | An observational post-marketing safety registry of Sativex | All ages (including children) | Anyone taking Sativex | Observational, prospective | Sativex (27 mg/mL THC, 25 mg/mL CBD) | CNP |
NCT02229032 | Genetic analysis between Charlotte’s web responders versus non-responders in a Dravet population | Children and adults (0–50) | Dravet syndrome | Observational, cohort, cross-sectional | Charlotte’s web strain of medical marijuana | CNP |
NCT04406948 | Study of safety and efficacy of MGCND00EP1 as an add-on treatment in children and adolescents with resistant epilepsies | Children (1–18) | Drug-resistant epilepsy | Interventional, randomized, parallel assignment, placebo controlled, double blind | MGCND00EP1 (cannabis oil with 20:1 CBD:THC ratio), up to 25 mg/kg/day | NYR |
NCT02332655 | Cannabidiol expanded-access study in medically refractory Sturge–Weber syndrome | Children and adults (0.083–45) | Refractory Sturge–Weber syndrome | Interventional, single group assignment, open label | Epidiolex 2 mg/kg/day up to 25 mg/kg/day | NYR |
NCT04485104 | Safety, pharmacokinetics, and exploratory efficacy assessment of adjunctive cannabidiol oral solution (GWP42003-P) compared with standard-of-care antiepileptic therapy in patients 1 to <12 months of age with tuberous sclerosis complex who experience inadequately controlled seizures | Infants (1–11 mo) | Tuberous sclerosis complex | Interventional, randomized, parallel assignment, open label | Epidiolex | NYR |
NCT02783092 | A double-blind trial to evaluate efficacy and safety of cannabidiol as an add-on therapy for treatment in refractory epilepsy | Children (2–18) | Refractory epilepsy | Interventional, randomized, double-blind, placebo-controlled | CBD in corn oil, 5–25 mg/kg/day | R |
NCT03848481 | CBDV vs. placebo in children and adults up to age 30 with Prader–Willi syndrome | Children and young adults (0–30) | Prader–Willi syndrome | Interventional, randomized, parallel assignment, placebo controlled, double blind | CBDV (cannabidivarin) compound at 10 mg/kg/day, contains <0.2% THC | R |
NCT04611438 | Research on cognitive effects of cannabidiol on Dravet syndrome and Lennox–Gastaut syndrome | Children (2–18) | Lennox–Gastaut syndrome, Dravet syndrome | Interventional, single group assignment, open label | Cannabidiol 5 mg/kg/day up to 10 mg/kg/day | R |
NCT03196466 | Population pharmacokinetics of antiepileptic in pediatrics (EPIPOP) | Children (0–18) | Epilepsy | Observational, cohort, retrospective | CBD | R |
NCT03052738 | Medical marijuana in the pediatric central nervous system tumor population | Children (2–18) | Central nervous system tumor | Observational, cohort, prospective | Medical marijuana | R |
NCT03848832 | Efficacy and Safety of Cannabidiol Oral Solution (GWP42003-P, CBD-OS) in Patients with Rett Syndrome (ARCH) | Children (2–18) | Rett Syndrome | Interventional, randomized, parallel assignment, double blind | Epidiolex 100 mg/mL twice a day | ANR |
NCT00153192 | Study to Evaluate the Efficacy of Dronabinol (Marinol) as an Add-On Therapy for Patients on Opioids for Chronic Pain | Anyone (includes children) | Chronic pain average score 4/10 or more | Interventional, randomized, crossover assignment, double blind | N/A | CNP |
NCT03614663 | Clinical Study of caNNabidiol in childrEn and adolesCenTs With Fragile X (CONNECT-FX) | Children (3–17) | Fragile X Syndrome (FMR1 full mutation) | Interventional, randomized, parallel assignment, double blind | ZYN002—CBD clear transdermal gel; patients up to 35 kg get 125 mg CBD, above that get 250 mg CBD | CNP |
NCT03824405 | Study of the Safety, Tolerability, and Efficacy of BTX 1204 in Patients with Moderate Atopic Dermatitis | Children and adults (12–70) | Atopic Dermatitis | Interventional, randomized, parallel assignment, double blind | BTX 1204 (synthetic cannabinoid) | CNP |
NCT03573518 | Evaluation of BTX 1503 in Patients with Moderate to Severe Acne Vulgaris | Children and adults (12–40) | Acne Vulgaris | Interventional, randomized, parallel assignment, double blind | BTX 1503 (synthetic cannabinoid) | CNP |
NCT03699527 | Medical Cannabis Registry and Pharmacology (Med Can Autism) | Children and young adults (1–21) | Autism | Observational, cohort, prospective | Any cannabis product | CNP |
NCT04252586 | An Open-Label Extension Study of Cannabidiol Oral Solution (GWP42003-P, CBD-OS) in Patients with Rett Syndrome (ARCH) | Children (2–18) | Have completed RCT GWND18064 (NCT02848832) | Interventional, single group assignment, open label | Epidiolex 100 mg/mL twice a day | EBI |
NCT03734731 | Cannabis vs. Opioid Pain Management: Objective Testing Comparisons | Children and adults (0–90) | Pain | Interventional, single group assignment, open label | Any cannabis product | NYR |
NCT04520685 | CAnnabidiol Study in Children with Autism Spectrum DisordEr (CASCADE) | Children (5–17) | Autism Spectrum Disorder | Interventional, randomized, crossover assignment, double blind | CBD up to 10 mg/kg/day | NYR |
NCT04721691 | Efficacy of Epidiolex in Patients with Electrical Status Epilepticus of Sleep (ESES) | Children (2–17) | Electrical status epilepticus of sleep | Interventional, randomized, crossover assignment, double blind | Epidiolex up to 20 mg/kg/day | NYR |
NCT04517799 | Trial of Cannabidiol to Treat Severe Behavior Problems in Children With Autism | Male children (7–14) | Autism | Interventional, randomized, crossover assignment, double blind | Epidiolex up to 20 mg/kg/day | R |
NCT03944447 | Outcomes Mandate National Integration with Cannabis as Medicine for Prevention and Treatment of COVID-19 (OMNI-Can) | Children and adults (7+) | Clinical diagnosis of a qualifying condition for medical marijuana | Interventional, non-randomized, single group assignment, open label | Medical cannabis, must be legal in patient’s state | R |
NCT03866941 | Acute and Chronic Toxicity of Some Synthetic Cannabinoids in Assiut Psychiatric Hospitals | Anyone (includes children) | Admitted to psychiatric facility for cannabinoid toxicity | Observational, prospective | Any cannabis product | R |
NCT03900923 | Cannabidiol for ASD Open Trial | Children (7–17) | Autism Spectrum Disorder | Interventional, single group assignment, open label | CBD 3, 6, or 9 mg/kg/day | R |
NCT03802799 | Open-Label Extension to Assess the Long-Term Safety and Tolerability of ZYN002 in Children and Adolescents with FXS | Children (3–18) | Have completed NCT03614663 | Interventional, single group assignment, open label | ZYN002—CBD clear transdermal gel | R |
NCT04447846 | Novel Cognitive Treatment Targets for Epidiolex in Sturge–Weber Syndrome | Children and adults (3–50) | Sturge–Weber syndrome | Interventional, single group assignment, open label | Epidiolex up to 20 mg/kg/day | R |
NCT04634136 | Full-Spectrum Medical Cannabis for Treatment of Spasticity in Patients with Severe Forms of Cerebral Palsy (HemPhar) | Children and young adults (5–25) | Cerebral palsy with spasticity | Interventional, randomized, crossover assignment, double blind | HemPhar cannabis extract THC:CBD 1:10 | R |
Cancer Type | Cannabinoid Used/ECS Component Investigated | Experiment Details | Proposed Mechanism | Ref |
---|---|---|---|---|
Pediatric low-grade glioma | CB1 receptor levels | Tumor samples from 33 patients | High CNR1 levels seen at diagnosis in tumors that remained stable or underwent spontaneous involution, implying that CB1 receptor activation may participate in this phenomenon. | [90] |
Brain tumors | CB2 receptor levels | Tumor samples from 20 adult patients and 25 pediatric patients | CB2 levels are higher in more malignant tumors and in areas of tumors with very proliferative/invasive cells. | [91] |
Neuroblastoma | CBD | SK-N-SH | CBD reduces proliferation, induces apoptosis, and increases caspase-3 levels of SK-N-SH cells in vitro and in vivo. | [92] |
Neuroblastoma | CBD | SH-SY5Y IMR32 | CBD altered expression of many miRNAs, including upregulation of has-let-7a and downregulation of has-miRNA-1972. These were confirmed to be responsible for increases in casp-3 and GAS-7 (by has-let-7a) and decreases in BCL2L1, SIRT1, and MYCN (by has-miRNA-1972). CBD also decreased AKT1 and increased PTEN, caused apoptosis, inhibited cell migration, and shifted the cellular metabolism towards glycolysis. | [93] |
Neuroblastoma | AM404 | SK-N-SH | AM404 inhibits the transcriptional activity of both NFAT and NF-κB through CB1 and TRPV1-independent pathways. AM404 also inhibits invasion and decreases expression of MMP-1, 3, and 7. | [94] |
Leukemia | CBD | Jurkat, EL4 (mouse lymphoma), MOLT-4 (adult ALL) | CBD decreases cell viability and increases apoptosis, both in vitro and in vivo. In vitro only, CBD increased PARP and caspase-3 cleavage, decreased mitochondrial membrane potential, and increased ROS, all of which were dependent on CB2 receptor activation. The CBD-induced decrease in p-p38 MAPK also depended on ROS production. | [95] |
Leukemia | THC | Jurkat cells | THC triggers extrinsic (cleavage of casp 8, 10) and intrinsic (mitochondrial membrane potential, cytochrome c release, cleavage of casp 3, 9) apoptotic pathways, but primarily the intrinsic pathway is responsible. | [96] |
Leukemia | THC | Jurkat cells | THC downregulated Raf-1/MEK/ERK signaling pathway by altering phosphorylation status, causing BAD to relocate to mitochondria and triggering intrinsic apoptosis. | [97] |
Leukemia | THC | Jurkat cells | THC induced apoptosis via CB2 receptor and increase in de novo ceramide production upstream of the intrinsic apoptotic pathway (mitochondrial hyperpolarization and cytochrome c release). | [98] |
Leukemia | CPP95540 | Jurkat cells Patient samples (T-ALL) | Apoptosis induced via production of ROS and activation of the mitochondrial intrinsic apoptosis pathway. This is not mediated via CB1 or CB2 receptors. This occurred both in an established cell line and in bone marrow samples from three T-ALL patients. | [99] |
Leukemia | CBD | Pediatric Leukemic lines: Jurkat (T-ALL), CCFR-CEM (T-ALL) Adult Leukemic lines: MOLT-3 (T-ALL), K562 (CML), Reh (B-ALL), and RS4;11 (B-ALL) | High concentrations of CBD inhibit migration and increases apoptosis and autophagy at high concentrations in T-ALL cells while low CBD concentrations increase proliferation. CBD directly increases mitochondrial Ca2+, which induces cytosolic 2+ and leads to apoptosis. CBD also increases ROS levels and activates caspases 3 and 9. | [100] |
Chronic lymphocytic leukemia | AM251 ACEA JWH133 AM630 R-(+)-methanandamide CBD | Patient-derived cell cultures | High CNR1 expression was correlated with poorer outcomes. Treatment with CB1 and/or CB2 agonists and antagonists did not alter viability or invasion significantly in patient cells or normal peripheral blood lymphocytes | [55] |
Leukemia | CBD, CBG, THC | CEM (ALL, pediatric) HL60 (promyelocytic leukemia, adult) | Combinations of any 2/3 cannabinoids are better at reducing cell viability than any cannabinoid alone, particularly combinations with CBD, especially the CBD/THC combination. These combinations worked synergistically with vincristine and cytarabine, but the synergy was better when cells were treated with cytarabine or vincristine before cannabinoid treatment. | [43] |
Rhabdomyosarcoma | THC, HU-210, Met-F-AEA | Rh4, Rh28 (translocation positive rhabdomyosarcoma) RMS13, RD, MRC-5 (lung fibroblasts) | Cannabinoid treatment decreased viability of translocation-positive rhabdomyosarcoma (tposRMS) mediated through the CB1 receptor. This was via the Akt/ERK pathway and required the upregulation of p8. | [101] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Malach, M.; Kovalchuk, I.; Kovalchuk, O. Medical Cannabis in Pediatric Oncology: Friend or Foe? Pharmaceuticals 2022, 15, 359. https://doi.org/10.3390/ph15030359
Malach M, Kovalchuk I, Kovalchuk O. Medical Cannabis in Pediatric Oncology: Friend or Foe? Pharmaceuticals. 2022; 15(3):359. https://doi.org/10.3390/ph15030359
Chicago/Turabian StyleMalach, Megan, Igor Kovalchuk, and Olga Kovalchuk. 2022. "Medical Cannabis in Pediatric Oncology: Friend or Foe?" Pharmaceuticals 15, no. 3: 359. https://doi.org/10.3390/ph15030359
APA StyleMalach, M., Kovalchuk, I., & Kovalchuk, O. (2022). Medical Cannabis in Pediatric Oncology: Friend or Foe? Pharmaceuticals, 15(3), 359. https://doi.org/10.3390/ph15030359