A new ESI-LC/MS approach for comprehensive metabolic profiling of phytocannabinoids in Cannabis

Paula Berman, Kate Futoran, Gil M Lewitus, Dzmitry Mukha, Maya Benami, Tomer Shlomi, David Meiri, Paula Berman, Kate Futoran, Gil M Lewitus, Dzmitry Mukha, Maya Benami, Tomer Shlomi, David Meiri

Abstract

Most clinical studies of Cannabis today focus on the contents of two phytocannabinoids: (-)-Δ9-trans-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD), regardless of the fact that the plant contains over 100 additional phytocannabinoids whose therapeutic effects and interplay have not yet been fully elucidated. This narrow view of a complex Cannabis plant is insufficient to comprehend the medicinal and pharmacological effects of the whole plant. In this study we suggest a new ESI-LC/MS/MS approach to identify phytocannabinoids from 10 different subclasses, and comprehensively profile the identified compounds in diverse medical Cannabis plants. Overall, 94 phytocannabinoids were identified and used for profiling 36 of the most commonly used Cannabis plants prescribed to patients in Israel. In order to demonstrate the importance of comprehensive phytocannabinoid analysis before and throughout medical Cannabis clinical trials, treatments, or experiments, we evaluated the anticonvulsant effects of several equally high-CBD Cannabis extracts (50% w/w). We found that despite the similarity in CBD contents, not all Cannabis extracts produced the same effects. This study's approach for phytocannabinoid profiling can enable researchers and physicians to analyze the effects of specific Cannabis compositions and is therefore critical when performing biological, medical and pharmacological-based research using Cannabis.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Phytocannabinoid biosynthesis and degradation routes and products according to the literature–. The most prevalent acid components are presented for each type of phytocannabinoid.
Figure 2
Figure 2
Observed chromatographic and MS characteristics of phytocannabinoid standard materials. (A) Total ion current (TIC) chromatogram of the 13 available phytocannabinoid standards, and their MS/MS spectra according to (B) CBG, (C) Δ9-THC, (D) CBD, (E) CBC, (F) Δ8-THC, (G) CBL, (H) CBN, (I) CBGA, (J) Δ9-THCA, (K) CBDA, (L) Δ9-THCV, (M) CBDV, and (N) CBDVA. Fragmentation structures for (O) CBG, (P) Δ9-THC, (Q) CBD, (R) CBC, (S) Δ8-THC, (T) CBL, and (U) CBN were determined according to the observed MS/MS spectra of each phytocannabinoid. Values of m/z in this figure are presented as nominal masses to improve interpretation of spectra. Accurate masses for the main fragments appear in Supplementary Figs S3–S9. The blue and green highlights mark identical m/z fragments for the neutral and acid phytocannabinoids.
Figure 3
Figure 3
LC/MS TIC of (A) untreated and (B) decarboxylated high-Δ9-THCA; (C) untreated and (D) decarboxylated high-CBDA; and (E) partly decarboxylated high-CBGA Cannabis strains injected in the prepared concentration, with annotated phytocannabinoids identified according to analytical standards on the samples with the highest expression.
Figure 4
Figure 4
Phytocannabinoid profiling of 36 different medical Cannabis strains. The LC/MS concentrations of each phytocannabinoid were normalized to the highest value and compared per phytocannabinoid in a heat map. Strains were arranged according to increasing Δ9-THCA content (first line) and phytocannabinoids by subclasses.
Figure 5
Figure 5
Phytocannabinoid profiling of a specific high-CBD Cannabis strain that is used to treat children with epilepsy. (A) LC/MS differential analysis of four samples from the same strain with the same genetics, planted and harvested in the same way and at the same time, but grown at four different greenhouses. Two samples were extracted from each plant and prepared in three dilutions (1:9, 1:99, and 1:999  v/v Cannabis extract to ethanol). Samples were injected to the LC/MS in duplicates. The average LC-MS concentrations of each phytocannabinoid were normalized to the highest value and compared per phytocannabinoid in a heat map. Strains were arranged according to increasing Δ9-THCA content (first line) and phytocannabinoids were arranged by subclasses. Quantitative comparison of the absolute concentrations of (B) CBDA, (C) Δ9-THCA, (D) CBGA, (E) CBCA, (F) CBDVA, (G) Δ9-THCVA, (H) CBGVA and (I) CBCVA, show significant differences between Cannabis samples from the same strain as a result of growing conditions. All values are reported as mean ± SD (one-way ANOVA, Tukey HSD post hoc test *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001).
Figure 6
Figure 6
Effect of equally high CBD Cannabis strain extracts on pentylenetetrazol (PTZ)-induced convulsions. (A) Comparison of the LC/MS concentrations of the five Cannabis extracts that were used to evaluate the anticonvulsant effect of Cannabis extracts using the PTZ-induced convulsion test in mice. Values were color-coded according to order of magnitude of concentrations. All strains contained similar CBD contents (50% w/w) but differed in the concentrations and ratios of other phytocannabinoids. (B) The analyzed Cannabis extracts were found to reduce PTZ-induced incidence of tonic-clonic seizures and mortality. (C) Both Cann4 and Cann5 extracts significantly increased latency to first tonic-clonic seizures (F5,103 = 4.4, p = 0.001). Values are reported as mean ± SEM, *p < 0.05, ***p < 0.001 vs. a control group. Percent of protection against incidence of tonic seizures and subsequent death to PTZ were compared among groups using the χ2 test (p > 0.05 for both seizure protection and mortality protection).

References

    1. ElSohly MA, Slade D. Chemical constituents of marijuana: the complex mixture of natural cannabinoids. Life Sci. 2005;78:539–548. doi: 10.1016/j.lfs.2005.09.011.
    1. Kogan NM, Mechoulam R. Cannabinoids in health and disease. Dialogues Clin. Neurosci. 2007;9:413–430.
    1. Lukhele ST, Motadi LR. Cannabidiol rather than Cannabis sativa extracts inhibit cell growth and induce apoptosis in cervical cancer cells. BMC Complement. Altern. Med. 2016;16:335. doi: 10.1186/s12906-016-1280-0.
    1. Deng L, Ng L, Ozawa T, Stella N. Quantitative analyses of synergistic responses between cannabidiol and DNA-damaging agents on the proliferation and viability of glioblastoma and neural progenitor cells in culture. J. Pharmacol. Exp. Ther. 2017;360:215–224. doi: 10.1124/jpet.116.236968.
    1. Dana N, et al. Cannabis sativa extract reduces cytoskeletal associated proteins in breast cancer cell line. Int. J. Cancer. Manag. 2017;10:e5474. doi: 10.5812/ijcm.5474.
    1. Watt G, Karl T. In vivo evidence for therapeutic properties of cannabidiol (CBD) for Alzheimer’s disease. Front. Pharmacol. 2017;8:20. doi: 10.3389/fphar.2017.00020.
    1. Tzadok M, et al. CBD-enriched medical cannabis for intractable pediatric epilepsy: The current Israeli experience. Seizure. 2016;35:41–44. doi: 10.1016/j.seizure.2016.01.004.
    1. Crippa JA, Crippa AC, Hallak JE, Martín-Santos R, Zuardi AW. Δ9-THC intoxication by cannabidiol-enriched Cannabis extract in two children with refractory epilepsy: Full remission after switching to purified cannabidiol. Front. Pharmacol. 2016;7:359. doi: 10.3389/fphar.2016.00359.
    1. Holgado MA, Martín-Banderas L, Álvarez-Fuentes J, Fernández-Arévalo M. Neuroprotective effect of cannabinoids nanoplatforms in neurodegenerative diseases. J. Drug Deliv. Sci. Technol. 2017;42:84–93. doi: 10.1016/j.jddst.2017.04.023.
    1. Mannucci C, et al. Neurological aspects of medical use of cannabidiol. CNS Neurol. Disord. Drug Targets. 2017;16:541–553. doi: 10.2174/1871527316666170413114210.
    1. Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology. 2005;65:812–819. doi: 10.1212/01.wnl.0000176753.45410.8b.
    1. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bronner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561–564. doi: 10.1038/346561a0.
    1. Chakravarti B, Ravi J, Ganju RK. Cannabinoids as therapeutic agents in cancer: current status and future implications. Oncotarget. 2014;5:5852–5872. doi: 10.18632/oncotarget.2233.
    1. Alexander SPH, Mathie A, Peters JA. G Protein‐coupled receptors. Br. J. Pharmacol. 2011;164:5–113. doi: 10.1111/j.1476-5381.2011.01649_1.x.
    1. Ross RA. Anandamide and vanilloid TRPV1 receptors. Br. J. Pharmacol. 2003;140:790–801. doi: 10.1038/sj.bjp.0705467.
    1. Dobovišek L, Hojnik M, Ferk P. Overlapping molecular pathways between cannabinoid receptors type 1 and 2 and estrogens/androgens on the periphery and their involvement in the pathogenesis of common diseases (Review) Int. J. Mol. Med. 2016;38:1642–1651. doi: 10.3892/ijmm.2016.2779.
    1. Turner, S. E., Williams, C. M., Iversen, L. & Whalley, B. J. Molecular pharmacology of phytocannabinoids in Phytocannabinoids (eds. Kinghorn A. D. Falk H. Gibbons S. & Kobayashi J.) 61–101 (Springer International Publishing, Switzerland 2017).
    1. Morales, P., Hurst, D. P. & Reggio, P. H. Molecular targets of the phytocannabinoids: A complex picture in Phytocannabinoids (eds Kinghorn A. D. Falk H. Gibbons S. & Kobayashi J.) 103–131 (Springer International Publishing, Switzerland 2017).
    1. Hanuš LO, Meyer SM, Muñoz E, Taglialatela-Scafati O, Appendino G. Phytocannabinoids: a unified critical inventory. Nat. Prod. Rep. 2016;33:1357–1392. doi: 10.1039/C6NP00074F.
    1. ElSohly Mahmoud A., Gul Waseem. Handbook of Cannabis. 2014. Constituents of Cannabis Sativa; pp. 3–22.
    1. Flores-Sanchez IJ, Verpoorte R. Secondary metabolism in Cannabis. Phytochem. Rev. 2008;7:615–639. doi: 10.1007/s11101-008-9094-4.
    1. Appendino G, Chianese G, Taglialatela-Scafati O. Cannabinoids: occurrence and medicinal chemistry. Curr. Med. Chem. 2011;18:1085–1099. doi: 10.2174/092986711794940888.
    1. Shani A, Mechoulam R. Cannabielsoic acids: Isolation and synthesis by a novel oxidative cyclization. Tetrahedron. 1974;30:2437–2446. doi: 10.1016/S0040-4020(01)97114-5.
    1. Crombie L, Ponsford R, Shani A, Yagnitinsky B, Mechoulam R. Hashish components. Photochemical production of cannabicyclol from cannabichromene. Tetrahedron Lett. 1968;9:5771–5772. doi: 10.1016/S0040-4039(00)76346-5.
    1. Shoyama Y, Oku R, Yamauchi T, Nishioka I. Cannabis. VI. Cannabicyclolic acid. Chem. Pharm. Bull. 1972;20:1927–1930. doi: 10.1248/cpb.20.1927.
    1. Brenneisen, R. Chemistry and analysis of phytocannabinoids and other Cannabis constituents in Marijuana and the Cannabinoids (ed. ElSohly, M. A.) 17–49 (Humana Press, Totowa, New Jersey, 2007).
    1. Turner CE, Elsohly MA. Constituents of Cannabis sativa L. XVI. A possible decomposition pathway of Δ9‐tetrahydrocannabinol to cannabinol. J. Heterocycl. Chem. 1979;16:1667–1668. doi: 10.1002/jhet.5570160834.
    1. Carbone M, et al. Chemical characterisation of oxidative degradation products of Δ9-THC. Tetrahedron. 2010;66:9497–9501. doi: 10.1016/j.tet.2010.10.025.
    1. Keller A, Leupin M, Mediavilla V, Wintermantel E. Influence of the growth stage of industrial hemp on chemical and physical properties of the fibres. Ind. Crop. Prod. 2001;13:35–48. doi: 10.1016/S0926-6690(00)00051-0.
    1. Raharjo TJ, Verpoorte R. Methods for the analysis of cannabinoids in biological materials: a review. Phytochem. Anal. 2004;15:79–94. doi: 10.1002/pca.753.
    1. Hazekamp A, Peltenburg A, Verpoorte R, Giroud C. Chromatographic and spectroscopic data of cannabinoids from Cannabis sativa L. J. Liq. Chromatogr. Relat. Technol. 2005;28:2361–2382. doi: 10.1080/10826070500187558.
    1. De Backer B, et al. Innovative development and validation of an HPLC/DAD method for the qualitative and quantitative determination of major cannabinoids in cannabis plant material. J. Chromatogr. B. 2009;877:4115–4124. doi: 10.1016/j.jchromb.2009.11.004.
    1. Fischedick JT, Hazekamp A, Erkelens T, Choi YH, Verpoorte R. Metabolic fingerprinting of Cannabis sativa L., cannabinoids and terpenoids for chemotaxonomic and drug standardization purposes. Phytochemistry. 2010;71:2058–2073. doi: 10.1016/j.phytochem.2010.10.001.
    1. Peschel W, Politi M. 1H NMR and HPLC/DAD for Cannabis sativa L. chemotype distinction, extract profiling and specification. Talanta. 2015;140:150–165. doi: 10.1016/j.talanta.2015.02.040.
    1. Giese MW, Lewis MA, Giese L, Smith KM. Method for the analysis of cannabinoids and terpenes in Cannabis. J. AOAC Int. 2015;98:1503–1522. doi: 10.5740/jaoacint.15-116.
    1. Hazekamp A, Tejkalová K, Papadimitriou S. Cannabis: from cultivar to chemovar II—a metabolomics approach to Cannabis classification. Cannabis Cannabinoid Res. 2016;1:202–215. doi: 10.1089/can.2016.0017.
    1. Aizpurua-Olaizola O, et al. Evolution of the cannabinoid and terpene content during the growth of Cannabis sativa plants from different chemotypes. J. Nat. Prod. 2016;79:324–331. doi: 10.1021/acs.jnatprod.5b00949.
    1. Vergara D, et al. Compromised external validity: Federally produced Cannabis does not reflect legal markets. Sci. Rep. 2017;7:46528. doi: 10.1038/srep46528.
    1. Thomas BF, Compton DR, Martin BR. Characterization of the lipophilicity of natural and synthetic analogs of Δ9-tetrahydrocannabinol and its relationship to pharmacological potency. J. Pharmacol. Exp. Ther. 1990;255:624–630.
    1. Vree TB. Mass spectrometry of cannabinoids. J. Pharm. Sci. 1977;66:1444–1450. doi: 10.1002/jps.2600661025.
    1. Harvey DJ. Mass spectrometry of the cannabinoids and their metabolites. Mass Spectrom. Rev. 1987;6:135–229. doi: 10.1002/mas.1280060104.
    1. Kajima M, Piraux M. The biogenesis of cannabinoids in Cannabis sativa. Phytochemistry. 1982;21:67–69. doi: 10.1016/0031-9422(82)80016-2.
    1. Shoyama Y, Hirano H, Nishioka I. Biosynthesis of propyl cannabinoid acid and its biosynthetic relationship with pentyl and methyl cannabinoid acids. Phytochemistry. 1984;23:1909–1912. doi: 10.1016/S0031-9422(00)84939-0.
    1. Flores-Sanchez IJ, Linthorst HJ, Verpoorte R. In silicio expression analysis of PKS genes isolated from Cannabis sativa L. Genet. Mol. Biol. 2010;33:633–636. doi: 10.1590/S1415-47572010005000088.
    1. Sumner LW, et al. Proposed minimum reporting standards for chemical analysis. Metabolomics. 2007;3:211–221. doi: 10.1007/s11306-007-0082-2.
    1. Santos RG, Hallak JE, Leite JP, Zuardi AW, Crippa JA. Phytocannabinoids and epilepsy. J. Clin. Pharm. Ther. 2015;40:135–143. doi: 10.1111/jcpt.12235.
    1. Number of Legal Medical Marijuana Patients. (2018).
    1. Russo EB, Taming THC. potential cannabis synergy and phytocannabinoid-terpenoid entourage effects: Phytocannabinoid-terpenoid entourage effects. Br. J. Pharmacol. 2011;163:1344–1364. doi: 10.1111/j.1476-5381.2011.01238.x.
    1. Sanchez-Ramos J. The entourage effect of the phytocannabinoids. Ann. Neurol. 2015;77:1083. doi: 10.1002/ana.24402.
    1. Annesley TM. Ion suppression in mass spectrometry. Clin. Chem. 2003;49:1041–1044. doi: 10.1373/49.7.1041.
    1. Furey A, Moriarty M, Bane V, Kinsella B, Lehane M. Ion suppression; a critical review on causes, evaluation, prevention and applications. Talanta. 2013;115:104–122. doi: 10.1016/j.talanta.2013.03.048.
    1. Hazekamp, A. Cannabis; extracting the medicine. Department of Pharmacognosy (IBL), Faculty of Science, Leiden University (2007).
    1. Jones NA, et al. Cannabidiol displays antiepileptiform and antiseizure properties in vitro and in vivo. J. Pharmacol. Exp. Ther. 2010;332:569–577. doi: 10.1124/jpet.109.159145.
    1. Klein BD, et al. Evaluation of cannabidiol in animal seizure models by the epilepsy therapy screening program (ETSP) Neurochem. Res. 2017;42:1939–1948. doi: 10.1007/s11064-017-2287-8.

Source: PubMed

3
Se inscrever