Figure 1 - uploaded by Guillermo Velasco
Content may be subject to copyright.
Pancreatic tumor cells express cannabinoid receptors. A, total RNA was isolated from the corresponding cell lines and reverse transcription-PCR (RT-PCR) using selective primers for human CB 1 , CB 2 , or GAPDH was done. Representative of two experiments. P, Panc1; M, MiaPaCa2; B, BxPc3; C, Capan2. B, cell lysates were obtained from the corresponding cell lines and CB 1 , CB 2 , and a -tubulin protein levels were determined by Western blot. Spleen ( Sp ) was used as a positive control for CB 1 and CB 2 expression. Representative of two experiments. C, representative (63 Â ) images of CB receptor–stained 

Pancreatic tumor cells express cannabinoid receptors. A, total RNA was isolated from the corresponding cell lines and reverse transcription-PCR (RT-PCR) using selective primers for human CB 1 , CB 2 , or GAPDH was done. Representative of two experiments. P, Panc1; M, MiaPaCa2; B, BxPc3; C, Capan2. B, cell lysates were obtained from the corresponding cell lines and CB 1 , CB 2 , and a -tubulin protein levels were determined by Western blot. Spleen ( Sp ) was used as a positive control for CB 1 and CB 2 expression. Representative of two experiments. C, representative (63 Â ) images of CB receptor–stained 

Source publication
Article
Full-text available
Pancreatic adenocarcinomas are among the most malignant forms of cancer and, therefore, it is of especial interest to set new strategies aimed at improving the prognostic of this deadly disease. The present study was undertaken to investigate the action of cannabinoids, a new family of potential antitumoral agents, in pancreatic cancer. We show tha...

Contexts in source publication

Context 1
... of pancreatic tumor cells in vitro. We investigated the effect of cannabinoids on pancreatic tumor cells. First, we determined the expression of cannabinoid receptors in four different human pancreatic tumor cell lines as well as in biopsies of human pancreatic tumors. CB 1 and CB 2 cannabinoid receptor mRNA ( Fig. 1 A ) and protein (Fig. 1 B and C ) were expressed in Panc1, MiaPaCa2, Capan2, and BxPc3 cell lines. In addition, mRNA for cannabinoid receptors was expressed in several human pancreatic tumor biopsies, whereas in samples obtained from normal pancreatic tissue, mRNA levels for these receptors were very low or could not be detected (Fig. 1 D ). This difference between tumor and nontransformed pancreatic tissue was further con- firmed by immunofluorescence analysis of CB receptors both in human biopsies from pancreatic cancer (Fig. 1 E ) and in pancreatic tumors generated in mice (Supplementary Fig. S1; see below). In both cases, expression of cannabinoid receptors was detected clearly in the tumor nodules but hardly in the surrounding pancreatic tissue. Next, we tested the effect of THC on cell viability. Incubation with THC led to a dose-dependent decrease in cell viability in the four lines tested (Fig. 2 A ). Because cells exhibited a different sensitivity to THC treatment, we chose MiaPaCa2 as the most sensitive line and Panc1 as a less sensitive line to confirm the involvement of cannabinoid receptors in THC antiproliferative action. Incubation with the CB 2 -selective antagonist SR144528, but not with the CB 1 -selective antagonist SR141716, prevented THC- induced loss of cell viability in both lines (Fig. 2 B and C ). Likewise, in MiaPaca2 and Panc1 cells, THC led to caspase-3 activation, a characteristic of apoptotic cell death, and preincubation with SR144528 abrogated this effect (Fig. 2 D and E ). Because de novo synthesized ceramide has been implicated in CB receptor–mediated apoptosis of glioma cells (19, 20), we tested the involvement of this pathway in our model. As shown in Supplementary Fig. S2 A , incubation with THC led to ceramide accumulation in MiaPaca2 and Panc1 cell lines, and preincubation with SR144528 or ISP-1, a selective inhibitor of serine palmitoyltransferase (the enzyme that catalyzes the rate-limiting step of ceramide biosynthesis; ref. 21), prevented this accumulation. In addition, ISP-1 prevented the THC-induced ( a ) decrease of cell viability (Fig. 2 B and C ) and ( b ) activation of caspase 3 (Supplementary Fig. S2 B and C ), indicating that de novo synthesized ceramide is involved in THC-induced apoptosis of pancreatic tumor cells. apoptosis of pancreatic tumor cells. p8 (also designated as candidate of metastasis 1) is a stress-regulated protein related to the architectural factor HMG-I/Y (22). This protein has been implicated in a number of functions including the induction of apoptosis of pancreatic tumor cells (23). In addition, it has been shown that ceramide treatment leads to p8 up-regulation (24) and we have very recently identified this protein as an essential mediator of cannabinoid antitumoral action in gliomas (25). We therefore tested the involvement of this protein in the antiproliferative effect of THC in our cells. p8 mRNA levels increased after THC treatment of MiaPaca2 cells, and incubation with SR144528 (Fig. 3 A ) or ISP-1 (Fig. 3 B ) prevented this effect. Moreover, knockdown of p8 mRNA using a selective siRNA prevented THC-induced apoptosis of MiaPaCa2 cells (Fig. 3 C ), confirming the implication of this gene in the response to THC in our model. Our next step was to identify genes downstream of p8 that could participate in the antitumoral effect of THC. By comparing the mRNA expression profiles of p8-deficient fibroblasts and fibroblasts with enforced p8 expression, several p8-dependent genes have been implicated in apoptotic signaling (25). Among these genes, we selected the endoplasmic reticulum (ER) stress–related proteins ATF-4 (26, 27) and TRB3 (28) as potential mediators of p8- dependent effects in our cells. Incubation with THC led to a parallel increase in p8, ATF-4, and TRB3 mRNA levels, which was prevented by incubation with ISP-1 (Fig. 3 D ). In addition, knockdown of p8 mRNA prevented ATF-4 and TRB3 up-regulation (Fig. 3 E ). Moreover, knockdown of ATF-4 or TRB3 mRNA also prevented THC-induced apoptosis (Fig. 3 F ). Taken together, these observations support that challenge with THC triggers a ceramide- and p8-controlled apoptotic response in pancreatic tumor cells, which involves up-regulation of these genes. models in vivo . To evaluate the antiproliferative effect of cannabinoids on pancreatic tumors in vivo , we first generated tumor xenografts by s.c. injection of MiaPaCa2 cells in immunodeficient mice. As shown in Fig. 4, peritumoral treatment with THC or the CB 2 -selective (and therefore psychoactivity devoid; ref. 19) cannabinoid agonist JWH-133 reduced notably the growth of the established pancreatic tumors. Next, we generated tumors by intrapancreatic injection of MiaPaCa2 cells to investigate the antitumoral effect of cannabinoids in a model that resembles more closely the niche of pancreatic cancer spreading. The synthetic cannabinoid agonist WIN 55,212-2 was selected for i.p. administration owing to its better bioavailability than THC and other classic cannabinoids. Experiments were done to confirm that this compound, which exhibits affinity for both cannabinoid receptor types (7), induces apoptosis of pancreatic cancer cells via the same endoplasmic reticulum stress–related proapoptotic pathway as THC (Supplementary Fig. S3). Administration of ...
Context 2
... of pancreatic tumor cells in vitro. We investigated the effect of cannabinoids on pancreatic tumor cells. First, we determined the expression of cannabinoid receptors in four different human pancreatic tumor cell lines as well as in biopsies of human pancreatic tumors. CB 1 and CB 2 cannabinoid receptor mRNA ( Fig. 1 A ) and protein (Fig. 1 B and C ) were expressed in Panc1, MiaPaCa2, Capan2, and BxPc3 cell lines. In addition, mRNA for cannabinoid receptors was expressed in several human pancreatic tumor biopsies, whereas in samples obtained from normal pancreatic tissue, mRNA levels for these receptors were very low or could not be detected (Fig. 1 D ). This difference between tumor and nontransformed pancreatic tissue was further con- firmed by immunofluorescence analysis of CB receptors both in human biopsies from pancreatic cancer (Fig. 1 E ) and in pancreatic tumors generated in mice (Supplementary Fig. S1; see below). In both cases, expression of cannabinoid receptors was detected clearly in the tumor nodules but hardly in the surrounding pancreatic tissue. Next, we tested the effect of THC on cell viability. Incubation with THC led to a dose-dependent decrease in cell viability in the four lines tested (Fig. 2 A ). Because cells exhibited a different sensitivity to THC treatment, we chose MiaPaCa2 as the most sensitive line and Panc1 as a less sensitive line to confirm the involvement of cannabinoid receptors in THC antiproliferative action. Incubation with the CB 2 -selective antagonist SR144528, but not with the CB 1 -selective antagonist SR141716, prevented THC- induced loss of cell viability in both lines (Fig. 2 B and C ). Likewise, in MiaPaca2 and Panc1 cells, THC led to caspase-3 activation, a characteristic of apoptotic cell death, and preincubation with SR144528 abrogated this effect (Fig. 2 D and E ). Because de novo synthesized ceramide has been implicated in CB receptor–mediated apoptosis of glioma cells (19, 20), we tested the involvement of this pathway in our model. As shown in Supplementary Fig. S2 A , incubation with THC led to ceramide accumulation in MiaPaca2 and Panc1 cell lines, and preincubation with SR144528 or ISP-1, a selective inhibitor of serine palmitoyltransferase (the enzyme that catalyzes the rate-limiting step of ceramide biosynthesis; ref. 21), prevented this accumulation. In addition, ISP-1 prevented the THC-induced ( a ) decrease of cell viability (Fig. 2 B and C ) and ( b ) activation of caspase 3 (Supplementary Fig. S2 B and C ), indicating that de novo synthesized ceramide is involved in THC-induced apoptosis of pancreatic tumor cells. apoptosis of pancreatic tumor cells. p8 (also designated as candidate of metastasis 1) is a stress-regulated protein related to the architectural factor HMG-I/Y (22). This protein has been implicated in a number of functions including the induction of apoptosis of pancreatic tumor cells (23). In addition, it has been shown that ceramide treatment leads to p8 up-regulation (24) and we have very recently identified this protein as an essential mediator of cannabinoid antitumoral action in gliomas (25). We therefore tested the involvement of this protein in the antiproliferative effect of THC in our cells. p8 mRNA levels increased after THC treatment of MiaPaca2 cells, and incubation with SR144528 (Fig. 3 A ) or ISP-1 (Fig. 3 B ) prevented this effect. Moreover, knockdown of p8 mRNA using a selective siRNA prevented THC-induced apoptosis of MiaPaCa2 cells (Fig. 3 C ), confirming the implication of this gene in the response to THC in our model. Our next step was to identify genes downstream of p8 that could participate in the antitumoral effect of THC. By comparing the mRNA expression profiles of p8-deficient fibroblasts and fibroblasts with enforced p8 expression, several p8-dependent genes have been implicated in apoptotic signaling (25). Among these genes, we selected the endoplasmic reticulum (ER) stress–related proteins ATF-4 (26, 27) and TRB3 (28) as potential mediators of p8- dependent effects in our cells. Incubation with THC led to a parallel increase in p8, ATF-4, and TRB3 mRNA levels, which was prevented by incubation with ISP-1 (Fig. 3 D ). In addition, knockdown of p8 mRNA prevented ATF-4 and TRB3 up-regulation (Fig. 3 E ). Moreover, knockdown of ATF-4 or TRB3 mRNA also prevented THC-induced apoptosis (Fig. 3 F ). Taken together, these observations support that challenge with THC triggers a ceramide- and p8-controlled apoptotic response in pancreatic tumor cells, which involves up-regulation of these genes. models in vivo . To evaluate the antiproliferative effect of cannabinoids on pancreatic tumors in vivo , we first generated tumor xenografts by s.c. injection of MiaPaCa2 cells in immunodeficient mice. As shown in Fig. 4, peritumoral treatment with THC or the CB 2 -selective (and therefore psychoactivity devoid; ref. 19) cannabinoid agonist JWH-133 reduced notably the growth of the established pancreatic tumors. Next, we generated tumors by intrapancreatic injection of MiaPaCa2 cells to investigate the antitumoral effect of cannabinoids in a model that resembles more closely the niche of pancreatic cancer spreading. The synthetic cannabinoid agonist WIN 55,212-2 was selected for i.p. administration owing to its better ...
Context 3
... of pancreatic tumor cells in vitro. We investigated the effect of cannabinoids on pancreatic tumor cells. First, we determined the expression of cannabinoid receptors in four different human pancreatic tumor cell lines as well as in biopsies of human pancreatic tumors. CB 1 and CB 2 cannabinoid receptor mRNA ( Fig. 1 A ) and protein (Fig. 1 B and C ) were expressed in Panc1, MiaPaCa2, Capan2, and BxPc3 cell lines. In addition, mRNA for cannabinoid receptors was expressed in several human pancreatic tumor biopsies, whereas in samples obtained from normal pancreatic tissue, mRNA levels for these receptors were very low or could not be detected (Fig. 1 D ). This difference between tumor and nontransformed pancreatic tissue was further con- firmed by immunofluorescence analysis of CB receptors both in human biopsies from pancreatic cancer (Fig. 1 E ) and in pancreatic tumors generated in mice (Supplementary Fig. S1; see below). In both cases, expression of cannabinoid receptors was detected clearly in the tumor nodules but hardly in the surrounding pancreatic tissue. Next, we tested the effect of THC on cell viability. Incubation with THC led to a dose-dependent decrease in cell viability in the four lines tested (Fig. 2 A ). Because cells exhibited a different sensitivity to THC treatment, we chose MiaPaCa2 as the most sensitive line and Panc1 as a less sensitive line to confirm the involvement of cannabinoid receptors in THC antiproliferative action. Incubation with the CB 2 -selective antagonist SR144528, but not with the CB 1 -selective antagonist SR141716, prevented THC- induced loss of cell viability in both lines (Fig. 2 B and C ). Likewise, in MiaPaca2 and Panc1 cells, THC led to caspase-3 activation, a characteristic of apoptotic cell death, and preincubation with SR144528 abrogated this effect (Fig. 2 D and E ). Because de novo synthesized ceramide has been implicated in CB receptor–mediated apoptosis of glioma cells (19, 20), we tested the involvement of this pathway in our model. As shown in Supplementary Fig. S2 A , incubation with THC led to ceramide accumulation in MiaPaca2 and Panc1 cell lines, and preincubation with SR144528 or ISP-1, a selective inhibitor of serine palmitoyltransferase (the enzyme that catalyzes the rate-limiting step of ceramide biosynthesis; ref. 21), prevented this accumulation. In addition, ISP-1 prevented the THC-induced ( a ) decrease of cell viability (Fig. 2 B and C ) and ( b ) activation of caspase 3 (Supplementary Fig. S2 B and C ), indicating that de novo synthesized ceramide is involved in THC-induced apoptosis of pancreatic tumor cells. apoptosis of pancreatic tumor cells. p8 (also designated as candidate of metastasis 1) is a stress-regulated protein related to the architectural factor HMG-I/Y (22). This protein has been implicated in a number of functions including the induction of apoptosis of pancreatic tumor cells (23). In addition, it has been shown that ceramide treatment leads to p8 up-regulation (24) and we have very recently identified this protein as an essential mediator of cannabinoid antitumoral action in gliomas (25). We therefore tested the involvement of this protein in the antiproliferative effect of THC in our cells. p8 mRNA levels increased after THC treatment of MiaPaca2 cells, and incubation with SR144528 (Fig. 3 A ) or ISP-1 (Fig. 3 B ) prevented this effect. Moreover, knockdown of p8 mRNA using a selective siRNA prevented THC-induced apoptosis of MiaPaCa2 cells (Fig. 3 C ), confirming the implication of this gene in the response to THC in our model. Our next step was to identify genes downstream of p8 that could participate in the antitumoral effect of THC. By comparing the mRNA expression profiles of p8-deficient fibroblasts and fibroblasts with enforced p8 expression, several p8-dependent genes have been implicated in apoptotic signaling (25). Among these genes, we selected the endoplasmic reticulum (ER) stress–related proteins ATF-4 (26, 27) and TRB3 (28) as potential mediators of p8- dependent effects in our cells. Incubation with THC led to a parallel increase in p8, ATF-4, and TRB3 mRNA levels, which was prevented by incubation with ISP-1 (Fig. 3 D ). In addition, knockdown of p8 mRNA prevented ATF-4 and TRB3 up-regulation (Fig. 3 E ). Moreover, knockdown of ATF-4 or TRB3 mRNA also prevented THC-induced apoptosis (Fig. 3 F ). Taken together, these observations support that challenge with THC triggers a ceramide- and p8-controlled apoptotic response in pancreatic tumor cells, which involves up-regulation of these genes. models in vivo . To evaluate the antiproliferative effect of cannabinoids on pancreatic tumors in vivo , we first generated tumor xenografts by s.c. injection of MiaPaCa2 cells in immunodeficient mice. As shown in Fig. 4, peritumoral treatment with THC or the CB 2 -selective (and therefore psychoactivity devoid; ref. 19) cannabinoid agonist JWH-133 reduced notably the growth of the established pancreatic tumors. Next, we generated tumors by intrapancreatic injection of MiaPaCa2 cells to investigate the antitumoral effect of cannabinoids in a model that resembles more closely the niche of pancreatic cancer spreading. The synthetic cannabinoid agonist WIN 55,212-2 was selected for i.p. administration owing to its better bioavailability than THC ...
Context 4
... of pancreatic tumor cells in vitro. We investigated the effect of cannabinoids on pancreatic tumor cells. First, we determined the expression of cannabinoid receptors in four different human pancreatic tumor cell lines as well as in biopsies of human pancreatic tumors. CB 1 and CB 2 cannabinoid receptor mRNA ( Fig. 1 A ) and protein (Fig. 1 B and C ) were expressed in Panc1, MiaPaCa2, Capan2, and BxPc3 cell lines. In addition, mRNA for cannabinoid receptors was expressed in several human pancreatic tumor biopsies, whereas in samples obtained from normal pancreatic tissue, mRNA levels for these receptors were very low or could not be detected (Fig. 1 D ). This difference between tumor and nontransformed pancreatic tissue was further con- firmed by immunofluorescence analysis of CB receptors both in human biopsies from pancreatic cancer (Fig. 1 E ) and in pancreatic tumors generated in mice (Supplementary Fig. S1; see below). In both cases, expression of cannabinoid receptors was detected clearly in the tumor nodules but hardly in the surrounding pancreatic tissue. Next, we tested the effect of THC on cell viability. Incubation with THC led to a dose-dependent decrease in cell viability in the four lines tested (Fig. 2 A ). Because cells exhibited a different sensitivity to THC treatment, we chose MiaPaCa2 as the most sensitive line and Panc1 as a less sensitive line to confirm the involvement of cannabinoid receptors in THC antiproliferative action. Incubation with the CB 2 -selective antagonist SR144528, but not with the CB 1 -selective antagonist SR141716, prevented THC- induced loss of cell viability in both lines (Fig. 2 B and C ). Likewise, in MiaPaca2 and Panc1 cells, THC led to caspase-3 activation, a characteristic of apoptotic cell death, and preincubation with SR144528 abrogated this effect (Fig. 2 D and E ). Because de novo synthesized ceramide has been implicated in CB receptor–mediated apoptosis of glioma cells (19, 20), we tested the involvement of this pathway in our model. As shown in Supplementary Fig. S2 A , incubation with THC led to ceramide accumulation in MiaPaca2 and Panc1 cell lines, and preincubation with SR144528 or ISP-1, a selective inhibitor of serine palmitoyltransferase (the enzyme that catalyzes the rate-limiting step of ceramide biosynthesis; ref. 21), prevented this accumulation. In addition, ISP-1 prevented the THC-induced ( a ) decrease of cell viability (Fig. 2 B and C ) and ( b ) activation of caspase 3 (Supplementary Fig. S2 B and C ), indicating that de novo synthesized ceramide is involved in THC-induced apoptosis of pancreatic tumor cells. apoptosis of pancreatic tumor cells. p8 (also designated as candidate of metastasis 1) is a stress-regulated protein related to the architectural factor HMG-I/Y (22). This protein has been implicated in a number of functions including the induction of apoptosis of pancreatic tumor cells (23). In addition, it has been shown that ceramide treatment leads to p8 up-regulation (24) and we have very recently identified this protein as an essential mediator of cannabinoid antitumoral action in gliomas (25). We therefore tested the involvement of this protein in the antiproliferative effect of THC in our cells. p8 mRNA levels increased after THC treatment of MiaPaca2 cells, and incubation with SR144528 (Fig. 3 A ) or ISP-1 (Fig. 3 B ) prevented this effect. Moreover, knockdown of p8 mRNA using a selective siRNA prevented THC-induced apoptosis of MiaPaCa2 cells (Fig. 3 C ), confirming the implication of this gene in the response to THC in our model. Our next step was to identify genes downstream of p8 that could participate in the antitumoral effect of THC. By comparing the mRNA expression profiles of p8-deficient fibroblasts and fibroblasts with enforced p8 expression, several p8-dependent genes have been implicated in apoptotic signaling (25). Among these genes, we selected the endoplasmic reticulum (ER) stress–related proteins ATF-4 (26, 27) and TRB3 (28) as potential mediators of p8- dependent effects in our cells. Incubation with THC led to a parallel increase in p8, ATF-4, and TRB3 mRNA levels, which was prevented by incubation with ISP-1 (Fig. 3 D ). In addition, knockdown of p8 mRNA prevented ATF-4 and TRB3 up-regulation (Fig. 3 E ). Moreover, knockdown of ATF-4 or TRB3 mRNA also prevented THC-induced apoptosis (Fig. 3 F ). Taken together, these observations support that challenge with THC triggers a ceramide- and p8-controlled apoptotic response in pancreatic tumor cells, which involves up-regulation of these genes. models in vivo . To evaluate the antiproliferative effect of cannabinoids on pancreatic tumors in vivo , we first generated tumor xenografts by s.c. injection of MiaPaCa2 cells in immunodeficient mice. As shown in Fig. 4, peritumoral treatment with THC or the CB 2 -selective (and therefore psychoactivity devoid; ref. 19) cannabinoid agonist JWH-133 reduced notably the growth of the established pancreatic tumors. Next, we generated tumors by intrapancreatic injection of MiaPaCa2 cells to investigate the antitumoral effect of cannabinoids in a model that resembles more closely the niche of pancreatic cancer spreading. The synthetic cannabinoid agonist WIN 55,212-2 was selected for i.p. administration owing to its better bioavailability than THC and other classic cannabinoids. Experiments were done to confirm that this compound, which exhibits affinity for both cannabinoid receptor types (7), induces apoptosis of pancreatic cancer cells via the same endoplasmic reticulum stress–related proapoptotic pathway as THC (Supplementary Fig. S3). Administration of WIN 55,212-2 not only remarkably reduced the growth of intrapancreatic tumors (Fig. 5 A ) but also significantly decreased the extension of the tumor cells to proximal (Fig. 5 B ) and distal (Fig. ...

Citations

... It has been observed that 2-AG suppresses pancreatic cancer cell proliferation and tumour growth in vitro and in vivo [129]; this 2-AG-induced antiproliferative effect is CB1-receptor dependent. Another receptor that obviously plays a role is GPR55 which is increased in human pancreatic ductal adenocarcinoma (PDAC) specimens [130]. ...
... They are summarised below (Table 12 below). In a mouse model of PDAC pharmacological blockade of GPR55 with CBD, GEM (a standard treatment), and CBD plus GEM increased the rodent lifespan compared to vehicle (mean survival 25.4 days, 27.8 days, 52.7 days, and 18.6 days respectively), with many of the signalling pathways involved in reducing PDAC cell cycle progression and cell growth identified [130]. Most interestingly, whereas pure THC, pure CBD, and pure GEM have demonstrated a benefit in terms of reduced tumour growth or longer survival, a study using an extract containing both cannabinoids, THC and CBD in a ratio of 1:6 did not demonstrate a significant impact on the tumour volume; even worth, the highest dose of 10 mg/kg extract showed a more pronounced tumour growth than that of the negative control [27] Overall, the best effect was achieved with a combination of high CBD and GEM. ...
Article
Full-text available
Amazingly, almost 50 years after the first demonstration of anticancer effects of cannabinoids in vitro and in vivo, well-designed clinical trials that definitively prove tumour-inhibiting effects in man are still missing. Whereas a large number of preclinical studies exist that describe tumour-inhibiting effects of cannabinoids, alone or in combination, but also in the form of medical cannabis or natural extracts in vitro, the number of in vivo studies is still limited. Even more limited are well-documented experiences in man. Most animal studies and experience with cannabinoids in man concern brain tumours. This review summarises the effects of phytocannabinoids in brain, breast, colorectal, head and neck, haematological, liver, lung, pancreatic, ovarian, prostate, and skin cancers in animal models and, if available, in patients. The large majority of animal studies demonstrate tumour-inhibiting effects of cannabinoids, thus confirming in vitro data. Experiences in cancer patients are almost exclusively limited to individual case reports and case series without a control group. Many questions are currently unanswered such as the role of pure cannabinoids compared to combinations, cannabinoids as the eventual sole cancer therapy, optimal dosages, or duration of treatment. Pure cannabidiol (CBD) seems to be superior to pure delta-9-tetrahydrocannabinol (THC) in experimental settings. The role of medical cannabis or extracts is less clear as they vary in their phytochemical composition. In conclusion, cannabis/cannabinoids may slow the progression of tumours. However, the hope that cannabinoids could eventually cure cancer as often spread in social media, is, at present, wishful thinking. Above all, well-designed clinical trials paired with long-term follow-up of cancer patients are needed.
... • THC-loaded nanoparticles exhibited significant cytotoxicity Martín-Banderas et al. (2015) • Increased tumor growth and reduced tumor immunogenicity Zhu et al. (2000) • Inhibited tumor growth and metastases Preet et al. (2008) • • Reduced proliferation, migration, invasion, and induced apoptosis for hepatocellular cancer (Leelawat et al., 2010) • Reduced expression of PDL-1, thereby enhancing immune checkpoint blockade of pancreatic cancer cells (Yang et al., 2020) • Decreased cell viability in pancreas cancer (Carracedo et al., 2006b) • Inhibits proliferation through Akt pathway in hepatocarcinoma (Rao et al., 2019) • Reduced cell proliferation, promoted apoptosis and elevated ROS levels in colon cancer (Aviello et al., 2012;Honarmand et al., 2018) • Cell cicle arrest in gastric cancer (Zhang et al., 2019) In vivo In vivo In vivo In vivo ...
... • AMPK-dependent activation of autophagy in hepatocellular carcinoma (Vara et al., 2011) • Reduced hepatocellular tumor growth (Vara et al., 2011) • Decreased metastasis and angiogenesis in colon cancer (Honarmand et al., 2018) • Regulation of tumor-immune microenvironment in pancreatic cancer (Qiu et al., 2019) • Reduced the growth of tpancreativ tumors (Carracedo et al., 2006b) • Reduced aberrant crypt foci polyps and tumor growth (Aviello et al., 2012;De Petrocellis et al., 2013;Honarmand et al., 2018;Jeong et al., 2019) Gynecological and urogenital cancers ...
Article
Full-text available
Cannabis enjoyed a “golden age” as a medicinal product in the late 19th, early 20th century, but the increased risk of overdose and abuse led to its criminalization. However, the 21st century have witnessed a resurgence of interest and a large body of literature regarding the benefits of cannabinoids have emerged. As legalization and decriminalization have spread around the world, cancer patients are increasingly interested in the potential utility of cannabinoids. Although eager to discuss cannabis use with their oncologist, patients often find them to be reluctant, mainly because clinicians are still not convinced by the existing evidence-based data to guide their treatment plans. Physicians should prescribe cannabis only if a careful explanation can be provided and follow up response evaluation ensured, making it mandatory for them to be up to date with the positive and also negative aspects of the cannabis in the case of cancer patients. Consequently, this article aims to bring some clarifications to clinicians regarding the sometimes-confusing various nomenclature under which this plant is mentioned, current legislation and the existing evidence (both preclinical and clinical) for the utility of cannabinoids in cancer patients, for either palliation of the associated symptoms or even the potential antitumor effects that cannabinoids may have.
... The two known CB receptors, CB1 and CB2, activate through G-protein-coupled receptors by inhibiting adenylate cyclase with a resultant decrease in cAMP levels [23]. The activated CB receptors also stimulate multiple signaling pathways through non-G protein pathways such as extracellular-signal-regulated kinase 1/2 (ERK1/2), Ca2+/calmodulin-dependent protein kinase kinase (CaMKK), phosphatidylinositol 3-kinase (PI3K/Akt) [32], Ceramide, and reactive oxygen species, all of which result in apoptosis and cell cycle arrest [32][33][34][35][36][37][38]. Other proposed mechanisms using CB receptor-independent signal transduction pathways include GPR55, TRPV1, TRPV2, and TRPMB [32][33][34][35][36][37][38]. ...
... The activated CB receptors also stimulate multiple signaling pathways through non-G protein pathways such as extracellular-signal-regulated kinase 1/2 (ERK1/2), Ca2+/calmodulin-dependent protein kinase kinase (CaMKK), phosphatidylinositol 3-kinase (PI3K/Akt) [32], Ceramide, and reactive oxygen species, all of which result in apoptosis and cell cycle arrest [32][33][34][35][36][37][38]. Other proposed mechanisms using CB receptor-independent signal transduction pathways include GPR55, TRPV1, TRPV2, and TRPMB [32][33][34][35][36][37][38]. ...
Article
Full-text available
Purpose: Cannabinoids (CBD) have anti-tumor activity against prostate cancer (PCa). Preclinical studies have demonstrated a significant decrease in prostate specific antigen (PSA) protein expression and reduced tumor growth in xenografts of LNCaP and DU-145 cells in athymic mice when treated with CBD. Over-the-counter CBD products may vary in activity without clear standardization, and Epidiolex is a standardized FDA-approved oral CBD solution for treatment of certain types of seizures. We aimed to assess the safety and preliminary anti-tumor activity of Epidiolex in patients with biochemically recurrent (BCR) PCa. Experimental design: This was an open-label, single center, phase I dose escalation study followed by a dose expansion in BCR patients after primary definitive local therapy (prostatectomy +/- salvage radiotherapy or primary definitive radiotherapy). Eligible patients were screened for urine tetrahydrocannabinol prior to enrollment. The starting dose level of Epidiolex was 600 mg by mouth once daily and escalated to 800 mg daily with the use of a Bayesian optimal interval design. All patients were treated for 90 days followed by a 10-day taper. The primary endpoints were safety and tolerability. Changes in PSA, testosterone levels, and patient-reported health-related quality of life were studied as secondary endpoints. Results: Seven patients were enrolled into the dose escalation cohort. There were no dose-limiting toxicities at the first two dose levels (600 mg and 800 mg). An additional 14 patients were enrolled at the 800 mg dose level into the dose expansion cohort. The most common adverse events were 55% diarrhea (grade 1-2), 25% nausea (grade 1-2), and 20% fatigue (grade 1-2). The mean PSA at baseline was 2.9 ng/mL. At the 12-week landmark time-point, 16 out of 18 (88%) had stable biochemical disease, one (5%) had partial biochemical response with the greatest measurable decline being 41%, and one (5%) had PSA progression. No statistically significant changes were observed in patient-reported outcomes (PROs), but PROs changed in the direction of supporting the tolerability of Epidiolex (e.g., emotional functioning improved). Conclusion: Epidiolex at a dose of 800 mg daily appears to be safe and tolerable in patients with BCR prostate cancer supporting a safe dose for future studies.
... In vitro studies report the potential antineoplastic effects of D 9 -tetrahydrocannabinol (D 9 -THC) and cannabidiol (CBD) on varying cancer cell lines. [1][2][3][4][5][6][7][8] Unfortunately, there are a considerable range of efficacies reported. In addition, other reports suggest that CBD and D 9 -THC have little or no efficacy. ...
... The in vitro anticancer activity of cannabinoids is artificially enhanced in the absence of serum As noted in the Introduction, there is considerable disagreement in the literature concerning the anticancer cytotoxic potential of the cannabinoids. [1][2][3][4][5][6][7][8][9][10][11][12][13] In reports that do report anticancer efficacy, FBS is often omitted or decreased during the drug incubation. To systematically address this issue, we investigated the anticancer activities of four different cannabinoids (CBD, D 9 -THC, KM-233, and HU-331; Fig. 1) against six different cancer cell lines in vitro (of three organ sites) (GBM-T98G and U87MG cells; melanoma-A375M and 1205Lu cells; and CRC-SW480 and HCT116 cells). ...
Article
Background: Studies have reported that cannabinoids, in particular Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD), significantly reduce cancer cell viability in vitro. Unfortunately, treatment conditions vary significantly across reports. In particular, a majority of reports utilize conditions with reduced serum concentrations (0-3%) that may compromise the growth of the cells themselves, as well as the observed results. Objectives: This study was designed to test the hypothesis that, based on their known protein binding characteristics, cannabinoids would be less effective in the presence of fetal bovine serum (FBS). Moreover, we wished to determine if the treatments served to be cytotoxic or cytostatic under these conditions. Methods: Six cancer cell lines, representing two independent lines of three different types of cancer (glioblastoma, melanoma, and colorectal cancer [CRC]), were treated with 10 μM pure Δ9-THC, CBD, KM-233, and HU-331 for 48 h (in the presence or absence of FBS). Cell viability was measured with the MTT assay. Dose-response curves were then generated comparing the potencies of the four cannabinoids under the same conditions. Results: We found that serum-free medium alone produces cell cycle arrest for CRC cells and slows cell growth for the other cancer types. The antineoplastic effects of three of the four cannabinoids (Δ9-THC, CBD, and KM-233) increase when serum is omitted from the media. In addition, dose-response curves for these drugs demonstrated lower IC50 values for serum-free media compared with the media with 10% serum in all cell lines. The fourth compound, HU-331, was equally effective under both conditions. A further confound we observed is that omission of serum produces dramatic binding of Δ9-THC and CBD to plastic. Conclusions: Treatment of cancer cells in the absence of FBS appears to enhance the potency of cannabinoids. However, omission of FBS itself compromises cell growth and represents a less physiological condition. Given the knowledge that cannabinoids are 90-95% protein bound and have well-known affinities for plastic, it may be ill-advised to treat cells under conditions where the cells are not growing optimally and where known concentrations cannot be assumed (i.e., FBS-free conditions).
... The main active substances found in cannabis plants are cannabidiol (CBD) and tetrahydrocannabinol (THC). [3][4][5][6] Even though, they have shown synergistic treatment effects in studies with the combination of CBD/synthetic cannabinoid receptor ligands and chemotherapy in xenograft and genetically modified spontaneous pancreatic cancer models, 7 no clinical studies to date showing treatment benefits of CBD or THC in patients with pancreatic cancer because of their weaker efficacy against the pancreatic cancer cells. [7][8][9][10] Recently, we have shown that the hydrogenated cannabinoids based on hexahydrocannabinol (HHC) and hexahydrocannabidiol (H4CBD) structures demonstrated a better potency than marketed poly(ADP-ribose) polymerase (PARP) inhibitors (Olaparib and Veliparib) and the parental molecules CBD and THC in human pancreatic ductal adenocarcinoma (PDAC) cell lines especially PANC1 and MiaPaCa2 thus, we believe that these new molecules, could serve as promising agents for better treatment outcome of patients diagnosed with pancreatic cancer. ...
Article
Full-text available
The characterization of any compound is important in the field of chemistry. As the field of cannabinoid chemistry grows so does the need for the characterization of new cannabinoids or rare cannabinoids that gain popularity within the consumer and research fields. Hexahydrocannabinol (HHC) a hydrogenated analogue of Δ9-tetrahydrocannabinol (THC), also found in trace amounts naturally within the Cannabis sativa plant, has been gaining attention and popularity within the cannabis industry. Hexahydrocannabidiol (H4CBD) is a synthetic hydrogenated analogue to cannabidiol (CBD). Identifying the diastereomers of the cannabinoids with instrumentation plays a huge role within the chemistry field adding valuable information of the structure and the parameters for others to identify such cannabinoids. Elucidation and characterization of HHC and H4CBD were performed using current analytical techniques such as 1D and 2D nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC), and gas chromatography-mass spectrometry (GC-MS), effectively characterizing both the diastereomers of HHC and H4CBD.
... Cannabidiol (CBD) is the most common cannabinoid in hemp and second most prevalent in the majority of cannabis cultivars, with a versatile pharmacological profile (Mechoulam 2005). Interestingly, studies have found that cannabinoids inhibit tumor cell growth and induce apoptosis in various cancer cells Blázquez et al. 2008;Guzmán et al. 2006;Carracedo et al. 2006;Javid et al. 2016;Blasco-Benito et al. 2018;Tomko et al. 2019). Despite the use of cannabis in the population and evidence of antitumoral activity by cannabinoids, little is known about the anticancer effects of cannabis use in bladder cancer. ...
Article
Full-text available
Introduction With the legalization of cannabis in multiple jurisdictions throughout the world, a larger proportion of the population consumes cannabis. Several studies have demonstrated anti-tumor effects of components present in cannabis in different models. Unfortunately, little is known about the potential anti-tumoral effects of cannabinoids in bladder cancer and how cannabinoids could potentially synergize with chemotherapeutic agents. Our study aims to identify whether a combination of cannabinoids, like cannabidiol and Δ ⁹ -tetrahydrocannabinol, with agents commonly used to treat bladder cancer, such as gemcitabine and cisplatin, can produce desirable synergistic effects. We also evaluated if co-treatment with different cannabinoids resulted in synergistic effects. Methods We generated concentration curves with several drugs, including several cannabinoids, to identify the range at which they could exert anti-tumor effects in bladder cancer cell lines. We tested the cytotoxic effects of gemcitabine (up to 100 nM), cisplatin (up to 100 μM), and cannabinoids (up to 10 μM) in T24 and TCCSUP cells. We also evaluated the activation of the apoptotic cascade and whether cannabinoids have the ability to reduce invasion in T24 cells. Results Cannabidiol, Δ ⁹ -tetrahydrocannabinol, cannabichromene, and cannabivarin reduce cell viability of bladder cancer cell lines, and their combination with gemcitabine or cisplatin may induce differential responses, from antagonistic to additive and synergistic effects, depending on the concentrations used. Cannabidiol and Δ ⁹ -tetrahydrocannabinol were also shown to induce apoptosis via caspase-3 cleavage and reduce invasion in a Matrigel assay. Cannabidiol and Δ ⁹ -tetrahydrocannabinol also display synergistic properties with other cannabinoids like cannabichromene or cannabivarin, although individual cannabinoids may be sufficient to reduce cell viability of bladder cancer cell lines. Discussion Our results indicate that cannabinoids can reduce human bladder transitional cell carcinoma cell viability, and that they can potentially exert synergistic effects when combined with other agents. Our in vitro results will form the basis for future studies in vivo and in clinical trials for the development of new therapies that could be beneficial for the treatment of bladder cancer in the future.
... Moreover, α-humulene and BCPO have no affinity to Frontiers in Chemistry frontiersin.org CB 1 and CB 2 , which explain that both compounds exhibited their biological activities through partially different mechanisms such as apoptosis induction, repression of the cell cycle, and inhibition of angiogenesis and metastasis (Carracedo et al., 2006). Many investigations have been performed to unravel the anticancer mechanism of BCPO, while that of BCP has hardly been studied (Fidyt et al., 2016). ...
Article
Full-text available
Introduction: Psidium cattleianum Sabine is a Brazilian native shrub cultivated for its edible fruit araçá (strawberry guava). P. cattleianum is recognized for health and food applications, although the essential oils (EOs) from the Egyptian inhabitant are not fully explored. The current study investigated the anti-inflammatory and cytotoxic activities of EOs from P. cattleianum leaves and flowers. Materials and methods: The EOs were obtained by three different methods viz ; the conventional hydro-distillation, microwave assisted hydro-distillation, and supercritical fluid extraction, while their analysis was accomplished using GC/MS. The derived EOs were screened for their anti-inflammatory activity in the 5-lipoxygenase, COX-1, and COX-2 enzyme based assays, while the anticancer potential was deduced from MTT cytotoxic assay, cell cycle, and western blotting analysis. Results and discussion: Among other methods, supercritical fluid extraction offered the highest EO yield, 0.62% (leaves) and 1.4% (flowers). GC/MS identified β-caryophyllene and α-humulene in both organs with high but variable percentages. The leaves demonstrated strong activity in inhibiting the 5-lipoxygenase enzyme (IC50 2.38), while the flowers, in inhibiting COX-2 (IC50 2.575). Moreover, the leaves showed potent, selective cytotoxicity to MCF-7 cells (IC50 5.32) via apoptosis by modulating the p53/Bax/Bcl2 axis. The deduced activities are possible due to the synergism between the volatile components that endorses P. cattleianum leaves’ EOs in the management of breast cancer and inflammatory disorders.
... The author demonstrated that Δ9-THC triggers the P8-regulated pathway and promotes apoptosis. Though this pathway is associated with autophagy, the authors do not discuss it [30]. Δ9-THC interacts with CB2, followed by inducing apoptosis to evoke breast cancer cell death, but the detailed molecular mechanism remains unexplored [31]. ...
Article
Full-text available
Tetrahydrocannabinols (THCs) antagonize the CB1 and CB2 cannabinoid receptors, whose signaling to the endocannabinoid system is essential for controlling cell survival and proliferation as well as psychoactive effects. Most tumor cells express a much higher level of CB1 and CB2; THCs have been investigated as potential cancer therapeutic due to their cannabimimetic properties. To date, THCs have been prescribed as palliative medicine to cancer patients but not as an anticancer modality. Growing evidence of preclinical research demonstrates that THCs reduce tumor progression by stimulating apoptosis and autophagy and inhibiting two significant hallmarks of cancer pathogenesis: metastasis and angiogenesis. However, the degree of their anticancer effects depends on the origin of the tumor site, the expression of cannabinoid receptors on tumor cells, and the dosages and types of THC. This review summarizes the current state of knowledge on the molecular processes that THCs target for their anticancer effects. It also emphasizes the substantial knowledge gaps that should be of concern in future studies. We also discuss the therapeutic effects of THCs and the problems that will need to be addressed in the future. Clarifying unanswered queries is a prerequisite to translating the THCs into an effective anticancer regime.
... We hypothesize that this might also be the reason why we did not observe significant changes in the p53 level, despite the observed pro-apoptotic effects of the analyzed compounds. Numerous reports show that CBD activates apoptosis-mediated cell death in several cancer cell lines, such as breast, colorectal, leukemia and pancreatic tumor cells; however, the reports concerning VSCC are scarce [40][41][42][43][44]. In addition, IBU and DIC were not intensively analyzed in regard to the treatment of VSCC cells. ...
Article
Full-text available
Vulvar squamous cell carcinoma (VSCC) is a rare malignancy with a relatively good prognosis. However, the prognosis remains poor for elderly patients and those with a significant depth of tumor invasion; thus, novel treatment modalities are needed. The aim of this study was to analyze the impact of cannabidiol (CBD) and its combination with NSAIDs, diclofenac (DIC) and ibuprofen (IBU) on VSCC cells. In this regard, the MTT test was applied for cytotoxicity analysis. Moreover, the influence of CBD, DIC and IBU, as well as their combinations, on apoptosis and cell cycle distribution were analyzed by flow cytometry. The mechanisms of action of the analyzed compounds, including their impact on NF-κB signaling, p53 and COX-2 expression were evaluated using Western blot. This study shows that CBD and its combinations with NSAIDs are cytotoxic to A431 cells, but they also reduce, in a dose-dependent manner, the viability of immortalized keratinocyte HaCaT cells, and human umbilical vein cell line, EA.hy926. Moreover, the compounds and their combinations induced apoptosis, diminished the NF-κB signaling activation and reduced COX-2 expression. We conclude that CBD and its combination with DIC or IBU are promising candidates for the adjuvant treatment of high-risk VSCC patients. However, their impact on non-cancerous cells requires careful evaluation.
... Over 100 phytocannabinoids have been identified (Mehmedic et al. 2010), but Δ 9 -tetrahydrocannabinol (THC) and cannabidiol (CBD) are the most common cannabinoids produced in the Cannabis plant (de Meijer et al. 2003;Mechoulam 2005). Interestingly, studies have found that multiple compounds from cannabis inhibit tumor cell growth and induce apoptosis in various cancer cells (Blázquez et al. , 2008Guzmán et al. 2006;Carracedo et al. 2006;Javid et al. 2016;Blasco-Benito et al. 2018;Tomko et al. 2019), but little is known about their effects in bladder cancer. Recently, a study suggested that cannabis-derived cannabichromene (CBC) and ∆ 9 -tetrahydrocannabinol displayed some synergy when used together in a model of urothelial cell carcinoma (Anis et al. 2021). ...
Article
Full-text available
Introduction Several studies have shown anti-tumor effects of components present in cannabis in different models. Unfortunately, little is known about the potential anti-tumoral effects of most compounds present in cannabis in bladder cancer and how these compounds could potentially positively or negatively impact the actions of chemotherapeutic agents. Our study aims to evaluate the effects of a compound found in Cannabis sativa that has not been extensively studied to date, cannflavin A, in bladder cancer cell lines. We aimed to identify whether cannflavin A co-treatment with agents commonly used to treat bladder cancer, such as gemcitabine and cisplatin, is able to produce synergistic effects. We also evaluated whether co-treatment of cannflavin A with various cannabinoids could produce synergistic effects. Methods Two transitional cell carcinoma cell lines were used to assess the cytotoxic effects of the flavonoid cannflavin A up to 100 μM. We tested the potential synergistic cytotoxic effects of cannflavin A with gemcitabine (up to 100 nM), cisplatin (up to 100 μM), and cannabinoids (up to 10 μM). We also evaluated the activation of the apoptotic cascade using annexin V and whether cannflavin A has the ability to reduce invasion using a Matrigel assay. Results Cell viability of bladder cancer cell lines was affected in a concentration-dependent fashion in response to cannflavin A, and its combination with gemcitabine or cisplatin induced differential responses—from antagonistic to additive—and synergism was also observed in some instances, depending on the concentrations and drugs used. Cannflavin A also activated apoptosis via caspase 3 cleavage and was able to reduce invasion by 50%. Interestingly, cannflavin A displayed synergistic properties with other cannabinoids like Δ ⁹ -tetrahydrocannabinol, cannabidiol, cannabichromene, and cannabivarin in the bladder cancer cell lines. Discussion Our results indicate that compounds from Cannabis sativa other than cannabinoids, like the flavonoid cannflavin A, can be cytotoxic to human bladder transitional carcinoma cells and that this compound can exert synergistic effects when combined with other agents. In vivo studies will be needed to confirm the activity of cannflavin A as a potential agent for bladder cancer treatment.