Mesenteric adipose–derived stromal cell lactoferrin may mediate protective effects in Crohn’s disease

Fat is not always bad
Article Type
Changed
Thu, 05/31/2018 - 16:52

Inflammatory bowel disease (IBD) and Crohn’s disease (CD), in particular, are characterized by an unusual ectopic extension of mesenteric adipose tissue. This intra-abdominal fat, also known as “creeping fat,” which wraps around the intestine during the onset of CD, is associated with inflammation and ulceration of the small or large intestine. The role of this fat in the development of CD, and whether it is protective or harmful, however, is not clear.

The current study demonstrates that adipose-derived stromal cells (ADSCs), the precursor cell population of adipose tissue, promote colonocyte proliferation and exhibit a differential gene expression profile in a disease-dependent manner. CD patient–derived ADSCs attenuated the severity of experimental colitis by releasing extracellular mediators, which exhibits a protective role for mesenteric adipose tissue during intestinal inflammation, according to Jill M. Hoffman, MD, and her colleagues at the University of California, Los Angeles. Increased expression and release of lactoferrin by ADSCs – an iron-binding glycoprotein and antimicrobial peptide usually found in large quantities in breast milk – was shown to be a likely mediator that could regulate inflammatory responses during CD. These results were published in Cellular and Molecular Gastroenterology and Hepatology (doi: 10.1016/j.jcmgh.2018.02.001).

Intestinal inflammation is primarily mediated by cytokine production, and targeted anticytokine therapy is the current standard for IBD treatment. The cytokine profile from CD patient–derived mesenteric ADSCs and fat tissue was significantly different from that of these patients’ disease-free counterparts. The authors hypothesized that mesenteric ADSCs release adipokines in response to disease-associated signals; this release of adipokines results from differential gene expression of mesenteric ADSCs in CD versus control patients. To test this hypothesis, conditioned media from CD patient–derived ADSCs was used to study gene expression in colonic intestinal epithelial cells in vitro and in mice with experimental colitis in vivo.

Using the Human LncRNA Expression Microarray V4.0, expression of 20,730 protein-coding mRNA targets was analysed, and 992 mRNA transcripts were found to be differentially (less than or equal to twofold change) expressed in CD patient–derived ADSCs, compared with control patient–derived ADSCs. Subsequent pathway analysis suggested activation of cellular growth and proliferation pathways with caspase 8 and p42/44 as top predicted networks that are differentially regulated in CD patient–derived ADSCs with respect to those of control patients.

The investigators treated intestinal epithelial cells – specifically, NCM460 – with conditioned 233 media from the same CD or control patient–derived ADSCs; subsequent microarray profiling using the GeneChip Human Gene ST Array showed increased expression of interleukin-17A, CCL23, and VEGFA. Ingenuity Pathway Analysis of mRNA expression indicated convergence in injury and inflammation pathways with the SERPINE1 gene, which suggests it’s the central regulator of the differential gene expression network.

In vivo, mice with active dextran sulfate sodium (DSS) colitis that were treated with daily injections of conditioned media from CD patients showed attenuation of colitis as compared with mice treated with vehicle or conditioned media from control patients. Furthermore, the mRNA expression of proinflammatory cytokines was reduced with increased proliferative response (as measured by Ki67 expression) in intestinal epithelial cells in the dextran sulfate sodium–treated mice receiving media from CD patients, compared with that in mice receiving media from control patients or vehicle-treated mice.

Cell proliferation was studied in real time (during a period of 120 hours) using the xCELLigence platform. The authors suggested that mesenteric adipose tissue–derived mediators may regulate proliferative responses in intestinal epithelial cells during intestinal inflammation, as observed by enhanced cell-doubling time in conditioned media from CD patient–derived ADSCs.

 

 


Levels of lactoferrin mRNA (validated by real time polymerase chain reaction; 92.70 ± 18.41 versus 28.98 ± 5.681; P less than .05) and protein (validated by ELISA; 142.2 ± 5.653 versus 120.1 ± 3.664; P less than .01) were increased in human mesenteric ADSCs and conditioned media from CD patients, respectively, compared with that from controls.

“Compared with mice receiving vehicle injections, mice receiving daily injections of lactoferrin had improved clinical scores (5.625 ± 0.565 versus 11.125 ± 0.743; n = 8) and colon length at day 7 (6.575 ± 0.1688 versus 5.613 ± 0.1445; n = 8). In addition, we found epithelial cell proliferation was increased in the colons of lactoferrin-treated mice with colitis, compared with vehicle-treated controls (3.548e7 ± 1.547e6 versus 1.184e7 ± 2.915e6; P less than .01),” said the authors.

Collectively, the presented data was suggestive of a protective role of mesenteric adipose tissue–derived mediators, such as lactoferrin, in the pathophysiology of CD.

The study was supported by the Broad Medical Research Program (IBD-0390), an NIDDK Q51856 Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship 1857 (F32 DK102322), the Neuroendocrine Assay and Models of Gastrointestinal Function and Disease Cores (P50 DK 64539), an AGA-1858 Broad Student Research Fellowship, the Blinder Center for Crohn’s 1859 Disease Research, the Eli and Edythe Broad Chair, and NIH/NIDDK grant DK047343.

The authors disclosed no conflicts of interest.

SOURCE: Hoffman J et al. Cell Molec Gastro Hepatol. doi: 10.1016/j.jcmgh.2018.02.001.

Body

 

Inflammatory bowel disease (IBD), including Crohn’s disease, is a chronic inflammatory condition of the gastrointestinal tract that is often associated with changes in adipose tissue. However, the pathophysiological significance of fat wrapping in Crohn’s disease remains largely elusive. A correlation of IBD with obesity has been established by a number of studies, which report 15%-40% of adults with IBD are obese. Obesity is found to have a negative effect on disease activity and progression to surgery in patients with Crohn’s disease. In contrast, adipose-derived stromal or stem cells exhibit regenerative and anti-inflammatory function.

A recent study published in Cellular and Molecular Gastroenterology and Hepatology by Jill M. Hoffman and her colleagues highlighted the immune-modulatory function of adipose-derived stromal cells (ADSCs) in Crohn’s disease patients. They observed that patient-derived ADSCs promote colonocyte proliferation and exhibit distinct gene expression patterns, compared with healthy controls. The authors successfully identified ADSC-derived lactoferrin, an iron binding glycoprotein and an antimicrobial peptide, as a potential immunoregulatory molecule.

Dr. Amlan Biswas
They concluded that the disease-dependent alterations in mesenteric adipose tissue–derived ADSCs might have a protective role in the pathophysiology of Crohn’s disease. The study elegantly highlighted the therapeutic potential of lactoferrin in IBD and provided new insight into the biology of ADSC.

Amlan Biswas, PhD, is an instructor in pediatrics at Harvard Medical School, Boston, and is affiliated with Boston Children’s Hospital in the division of gastroenterology and nutrition. He has no conflicts of interest

Publications
Topics
Sections
Body

 

Inflammatory bowel disease (IBD), including Crohn’s disease, is a chronic inflammatory condition of the gastrointestinal tract that is often associated with changes in adipose tissue. However, the pathophysiological significance of fat wrapping in Crohn’s disease remains largely elusive. A correlation of IBD with obesity has been established by a number of studies, which report 15%-40% of adults with IBD are obese. Obesity is found to have a negative effect on disease activity and progression to surgery in patients with Crohn’s disease. In contrast, adipose-derived stromal or stem cells exhibit regenerative and anti-inflammatory function.

A recent study published in Cellular and Molecular Gastroenterology and Hepatology by Jill M. Hoffman and her colleagues highlighted the immune-modulatory function of adipose-derived stromal cells (ADSCs) in Crohn’s disease patients. They observed that patient-derived ADSCs promote colonocyte proliferation and exhibit distinct gene expression patterns, compared with healthy controls. The authors successfully identified ADSC-derived lactoferrin, an iron binding glycoprotein and an antimicrobial peptide, as a potential immunoregulatory molecule.

Dr. Amlan Biswas
They concluded that the disease-dependent alterations in mesenteric adipose tissue–derived ADSCs might have a protective role in the pathophysiology of Crohn’s disease. The study elegantly highlighted the therapeutic potential of lactoferrin in IBD and provided new insight into the biology of ADSC.

Amlan Biswas, PhD, is an instructor in pediatrics at Harvard Medical School, Boston, and is affiliated with Boston Children’s Hospital in the division of gastroenterology and nutrition. He has no conflicts of interest

Body

 

Inflammatory bowel disease (IBD), including Crohn’s disease, is a chronic inflammatory condition of the gastrointestinal tract that is often associated with changes in adipose tissue. However, the pathophysiological significance of fat wrapping in Crohn’s disease remains largely elusive. A correlation of IBD with obesity has been established by a number of studies, which report 15%-40% of adults with IBD are obese. Obesity is found to have a negative effect on disease activity and progression to surgery in patients with Crohn’s disease. In contrast, adipose-derived stromal or stem cells exhibit regenerative and anti-inflammatory function.

A recent study published in Cellular and Molecular Gastroenterology and Hepatology by Jill M. Hoffman and her colleagues highlighted the immune-modulatory function of adipose-derived stromal cells (ADSCs) in Crohn’s disease patients. They observed that patient-derived ADSCs promote colonocyte proliferation and exhibit distinct gene expression patterns, compared with healthy controls. The authors successfully identified ADSC-derived lactoferrin, an iron binding glycoprotein and an antimicrobial peptide, as a potential immunoregulatory molecule.

Dr. Amlan Biswas
They concluded that the disease-dependent alterations in mesenteric adipose tissue–derived ADSCs might have a protective role in the pathophysiology of Crohn’s disease. The study elegantly highlighted the therapeutic potential of lactoferrin in IBD and provided new insight into the biology of ADSC.

Amlan Biswas, PhD, is an instructor in pediatrics at Harvard Medical School, Boston, and is affiliated with Boston Children’s Hospital in the division of gastroenterology and nutrition. He has no conflicts of interest

Title
Fat is not always bad
Fat is not always bad

Inflammatory bowel disease (IBD) and Crohn’s disease (CD), in particular, are characterized by an unusual ectopic extension of mesenteric adipose tissue. This intra-abdominal fat, also known as “creeping fat,” which wraps around the intestine during the onset of CD, is associated with inflammation and ulceration of the small or large intestine. The role of this fat in the development of CD, and whether it is protective or harmful, however, is not clear.

The current study demonstrates that adipose-derived stromal cells (ADSCs), the precursor cell population of adipose tissue, promote colonocyte proliferation and exhibit a differential gene expression profile in a disease-dependent manner. CD patient–derived ADSCs attenuated the severity of experimental colitis by releasing extracellular mediators, which exhibits a protective role for mesenteric adipose tissue during intestinal inflammation, according to Jill M. Hoffman, MD, and her colleagues at the University of California, Los Angeles. Increased expression and release of lactoferrin by ADSCs – an iron-binding glycoprotein and antimicrobial peptide usually found in large quantities in breast milk – was shown to be a likely mediator that could regulate inflammatory responses during CD. These results were published in Cellular and Molecular Gastroenterology and Hepatology (doi: 10.1016/j.jcmgh.2018.02.001).

Intestinal inflammation is primarily mediated by cytokine production, and targeted anticytokine therapy is the current standard for IBD treatment. The cytokine profile from CD patient–derived mesenteric ADSCs and fat tissue was significantly different from that of these patients’ disease-free counterparts. The authors hypothesized that mesenteric ADSCs release adipokines in response to disease-associated signals; this release of adipokines results from differential gene expression of mesenteric ADSCs in CD versus control patients. To test this hypothesis, conditioned media from CD patient–derived ADSCs was used to study gene expression in colonic intestinal epithelial cells in vitro and in mice with experimental colitis in vivo.

Using the Human LncRNA Expression Microarray V4.0, expression of 20,730 protein-coding mRNA targets was analysed, and 992 mRNA transcripts were found to be differentially (less than or equal to twofold change) expressed in CD patient–derived ADSCs, compared with control patient–derived ADSCs. Subsequent pathway analysis suggested activation of cellular growth and proliferation pathways with caspase 8 and p42/44 as top predicted networks that are differentially regulated in CD patient–derived ADSCs with respect to those of control patients.

The investigators treated intestinal epithelial cells – specifically, NCM460 – with conditioned 233 media from the same CD or control patient–derived ADSCs; subsequent microarray profiling using the GeneChip Human Gene ST Array showed increased expression of interleukin-17A, CCL23, and VEGFA. Ingenuity Pathway Analysis of mRNA expression indicated convergence in injury and inflammation pathways with the SERPINE1 gene, which suggests it’s the central regulator of the differential gene expression network.

In vivo, mice with active dextran sulfate sodium (DSS) colitis that were treated with daily injections of conditioned media from CD patients showed attenuation of colitis as compared with mice treated with vehicle or conditioned media from control patients. Furthermore, the mRNA expression of proinflammatory cytokines was reduced with increased proliferative response (as measured by Ki67 expression) in intestinal epithelial cells in the dextran sulfate sodium–treated mice receiving media from CD patients, compared with that in mice receiving media from control patients or vehicle-treated mice.

Cell proliferation was studied in real time (during a period of 120 hours) using the xCELLigence platform. The authors suggested that mesenteric adipose tissue–derived mediators may regulate proliferative responses in intestinal epithelial cells during intestinal inflammation, as observed by enhanced cell-doubling time in conditioned media from CD patient–derived ADSCs.

 

 


Levels of lactoferrin mRNA (validated by real time polymerase chain reaction; 92.70 ± 18.41 versus 28.98 ± 5.681; P less than .05) and protein (validated by ELISA; 142.2 ± 5.653 versus 120.1 ± 3.664; P less than .01) were increased in human mesenteric ADSCs and conditioned media from CD patients, respectively, compared with that from controls.

“Compared with mice receiving vehicle injections, mice receiving daily injections of lactoferrin had improved clinical scores (5.625 ± 0.565 versus 11.125 ± 0.743; n = 8) and colon length at day 7 (6.575 ± 0.1688 versus 5.613 ± 0.1445; n = 8). In addition, we found epithelial cell proliferation was increased in the colons of lactoferrin-treated mice with colitis, compared with vehicle-treated controls (3.548e7 ± 1.547e6 versus 1.184e7 ± 2.915e6; P less than .01),” said the authors.

Collectively, the presented data was suggestive of a protective role of mesenteric adipose tissue–derived mediators, such as lactoferrin, in the pathophysiology of CD.

The study was supported by the Broad Medical Research Program (IBD-0390), an NIDDK Q51856 Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship 1857 (F32 DK102322), the Neuroendocrine Assay and Models of Gastrointestinal Function and Disease Cores (P50 DK 64539), an AGA-1858 Broad Student Research Fellowship, the Blinder Center for Crohn’s 1859 Disease Research, the Eli and Edythe Broad Chair, and NIH/NIDDK grant DK047343.

The authors disclosed no conflicts of interest.

SOURCE: Hoffman J et al. Cell Molec Gastro Hepatol. doi: 10.1016/j.jcmgh.2018.02.001.

Inflammatory bowel disease (IBD) and Crohn’s disease (CD), in particular, are characterized by an unusual ectopic extension of mesenteric adipose tissue. This intra-abdominal fat, also known as “creeping fat,” which wraps around the intestine during the onset of CD, is associated with inflammation and ulceration of the small or large intestine. The role of this fat in the development of CD, and whether it is protective or harmful, however, is not clear.

The current study demonstrates that adipose-derived stromal cells (ADSCs), the precursor cell population of adipose tissue, promote colonocyte proliferation and exhibit a differential gene expression profile in a disease-dependent manner. CD patient–derived ADSCs attenuated the severity of experimental colitis by releasing extracellular mediators, which exhibits a protective role for mesenteric adipose tissue during intestinal inflammation, according to Jill M. Hoffman, MD, and her colleagues at the University of California, Los Angeles. Increased expression and release of lactoferrin by ADSCs – an iron-binding glycoprotein and antimicrobial peptide usually found in large quantities in breast milk – was shown to be a likely mediator that could regulate inflammatory responses during CD. These results were published in Cellular and Molecular Gastroenterology and Hepatology (doi: 10.1016/j.jcmgh.2018.02.001).

Intestinal inflammation is primarily mediated by cytokine production, and targeted anticytokine therapy is the current standard for IBD treatment. The cytokine profile from CD patient–derived mesenteric ADSCs and fat tissue was significantly different from that of these patients’ disease-free counterparts. The authors hypothesized that mesenteric ADSCs release adipokines in response to disease-associated signals; this release of adipokines results from differential gene expression of mesenteric ADSCs in CD versus control patients. To test this hypothesis, conditioned media from CD patient–derived ADSCs was used to study gene expression in colonic intestinal epithelial cells in vitro and in mice with experimental colitis in vivo.

Using the Human LncRNA Expression Microarray V4.0, expression of 20,730 protein-coding mRNA targets was analysed, and 992 mRNA transcripts were found to be differentially (less than or equal to twofold change) expressed in CD patient–derived ADSCs, compared with control patient–derived ADSCs. Subsequent pathway analysis suggested activation of cellular growth and proliferation pathways with caspase 8 and p42/44 as top predicted networks that are differentially regulated in CD patient–derived ADSCs with respect to those of control patients.

The investigators treated intestinal epithelial cells – specifically, NCM460 – with conditioned 233 media from the same CD or control patient–derived ADSCs; subsequent microarray profiling using the GeneChip Human Gene ST Array showed increased expression of interleukin-17A, CCL23, and VEGFA. Ingenuity Pathway Analysis of mRNA expression indicated convergence in injury and inflammation pathways with the SERPINE1 gene, which suggests it’s the central regulator of the differential gene expression network.

In vivo, mice with active dextran sulfate sodium (DSS) colitis that were treated with daily injections of conditioned media from CD patients showed attenuation of colitis as compared with mice treated with vehicle or conditioned media from control patients. Furthermore, the mRNA expression of proinflammatory cytokines was reduced with increased proliferative response (as measured by Ki67 expression) in intestinal epithelial cells in the dextran sulfate sodium–treated mice receiving media from CD patients, compared with that in mice receiving media from control patients or vehicle-treated mice.

Cell proliferation was studied in real time (during a period of 120 hours) using the xCELLigence platform. The authors suggested that mesenteric adipose tissue–derived mediators may regulate proliferative responses in intestinal epithelial cells during intestinal inflammation, as observed by enhanced cell-doubling time in conditioned media from CD patient–derived ADSCs.

 

 


Levels of lactoferrin mRNA (validated by real time polymerase chain reaction; 92.70 ± 18.41 versus 28.98 ± 5.681; P less than .05) and protein (validated by ELISA; 142.2 ± 5.653 versus 120.1 ± 3.664; P less than .01) were increased in human mesenteric ADSCs and conditioned media from CD patients, respectively, compared with that from controls.

“Compared with mice receiving vehicle injections, mice receiving daily injections of lactoferrin had improved clinical scores (5.625 ± 0.565 versus 11.125 ± 0.743; n = 8) and colon length at day 7 (6.575 ± 0.1688 versus 5.613 ± 0.1445; n = 8). In addition, we found epithelial cell proliferation was increased in the colons of lactoferrin-treated mice with colitis, compared with vehicle-treated controls (3.548e7 ± 1.547e6 versus 1.184e7 ± 2.915e6; P less than .01),” said the authors.

Collectively, the presented data was suggestive of a protective role of mesenteric adipose tissue–derived mediators, such as lactoferrin, in the pathophysiology of CD.

The study was supported by the Broad Medical Research Program (IBD-0390), an NIDDK Q51856 Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship 1857 (F32 DK102322), the Neuroendocrine Assay and Models of Gastrointestinal Function and Disease Cores (P50 DK 64539), an AGA-1858 Broad Student Research Fellowship, the Blinder Center for Crohn’s 1859 Disease Research, the Eli and Edythe Broad Chair, and NIH/NIDDK grant DK047343.

The authors disclosed no conflicts of interest.

SOURCE: Hoffman J et al. Cell Molec Gastro Hepatol. doi: 10.1016/j.jcmgh.2018.02.001.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CMGH

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Early results favor combo IL-15/anti-CD20 in indolent NHL

Article Type
Changed
Tue, 01/17/2023 - 11:16

 

– A combination of an immunostimulatory IL-15-based agent, ALT-803, with a therapeutic monoclonal antibody (mAb) against CD20, was well tolerated and had clinical activity in patients with indolent non-Hodgkin lymphoma (iNHL), according to preliminary findings from a phase 1 study.

“The cancer immunotherapy breakthrough that happened several years ago continues year after year, with a plethora of different modalities of immunotherapy at our disposal,” Todd A. Fehniger, MD, PhD, said at the annual meeting of the American Association for Cancer Research.

Immunotherapy with anti-CD20 mAbs, alone or in combination with chemotherapy, is a standard therapy for iNHL patients. Since iNHL cells express CD20, targeting it with mAbs triggers antitumor responses via cell surface receptors resulting in a potent antibody-dependent cellular toxicity. However, response in patients is highly heterogeneous, with relapse within a few months in a subset of patients. In addition, chemotherapeutic combinations can be toxic and result in serious and long-term complications.

“Relapsed or refractory iNHL is not curable and treatment strategies without long-term complications are needed,” said Dr. Fehniger, associate professor of medicine at Washington University, St. Louis.

In an attempt to address this, Dr. Fehniger and his colleagues combined rituximab, an anti-CD20 antibody, with a relatively new IL-15 agonist immunostimulatory agent called ALT-803.

In the phase 1 trial, the researchers enrolled patients with indolent non-Hodgkin lymphoma who had relapsed after at least 1 prior to CD20 antibody containing therapy. The study was a standard 3+3 dose escalation design with rituximab administered by intravenous infusion, 375 mg/m2 in four weekly doses, followed by a rest and four consolidation doses every 8 weeks for four cycles.

ALT-803 was administered concurrently at dose levels of 1 mcg/kg, 3 mcg/kg, and 6 mcg/kg IV followed by 6 mcg/kg, 10 mcg/kg, 15 mcg/kg, and 20 mcg/kg subcutaneously.

 

 


In total, 21 patients were treated: 16 patients had follicular lymphoma, four patients had marginal zone lymphoma, and one patient had small lymphocytic lymphoma. The median prior therapies received was two (range: 1-18) and five patients were treated who were refractory to prior anti-CD20 MAb therapy.

ALT-803 was well tolerated with no dose limiting toxicities or grade 4 or 5 adverse events. No patients discontinued ALT-803 and the recommended phase 2 dose was 20 mcg/kg subcutaneously. Grade 3 adverse events, regardless of attribution to ALT-803, included transient hypertension (14%), anemia (5%), nausea (5%), chills (5%), fever (5%), neutropenia (5%), and hyperglycemia (5%).

“Patients who received [subcutaneous] ALT-803 developed a unique injection site rash reaction that peaked 7-10 days later but resolved typically within 14 days. It was self-limited and resolved on its own,” Dr. Fehniger said.

At the time of the presentation, the best overall response rate was achieved in 11 of 21 patients (52%), with 9 complete responders (43%), and 2 partial responders (10%).
 

 


Of the 12 patients treated with ALT-803 subcutaneously, 11 patients had either stable disease, or partial or complete responses. All 11 patients remained on study and were in consolidation or follow-up and have not relapsed, Dr. Fehniger reported.

Among the five rituximab-refractory patients, the researchers observed one complete response, two patients with stable disease (45% and 36% tumor volume decrease), and two patients with partial disease. The durability of the responses can only be understood with longer follow-up, Dr. Fehniger said.

The peripheral blood of the patients was analyzed via flow cytometry and mass cytometry. Over the duration of four weekly doses, there was an increase in percentage (sixfold, P less than .001) and absolute number (10-fold, P less than .001) of natural killer cells at the 15-mcg/kg and 20-mcg/kg subcutaneous dose levels of ALT-803.

These results suggest that further studies of ALT-803 with other therapeutic targeting mAbs, or other immunotherapy modalities, are warranted, the researchers concluded.

Dr. Fehniger reported research funding from Altor BioScience.

SOURCE: Fehniger TA et al. AACR Annual Meeting, Abstract CT146.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– A combination of an immunostimulatory IL-15-based agent, ALT-803, with a therapeutic monoclonal antibody (mAb) against CD20, was well tolerated and had clinical activity in patients with indolent non-Hodgkin lymphoma (iNHL), according to preliminary findings from a phase 1 study.

“The cancer immunotherapy breakthrough that happened several years ago continues year after year, with a plethora of different modalities of immunotherapy at our disposal,” Todd A. Fehniger, MD, PhD, said at the annual meeting of the American Association for Cancer Research.

Immunotherapy with anti-CD20 mAbs, alone or in combination with chemotherapy, is a standard therapy for iNHL patients. Since iNHL cells express CD20, targeting it with mAbs triggers antitumor responses via cell surface receptors resulting in a potent antibody-dependent cellular toxicity. However, response in patients is highly heterogeneous, with relapse within a few months in a subset of patients. In addition, chemotherapeutic combinations can be toxic and result in serious and long-term complications.

“Relapsed or refractory iNHL is not curable and treatment strategies without long-term complications are needed,” said Dr. Fehniger, associate professor of medicine at Washington University, St. Louis.

In an attempt to address this, Dr. Fehniger and his colleagues combined rituximab, an anti-CD20 antibody, with a relatively new IL-15 agonist immunostimulatory agent called ALT-803.

In the phase 1 trial, the researchers enrolled patients with indolent non-Hodgkin lymphoma who had relapsed after at least 1 prior to CD20 antibody containing therapy. The study was a standard 3+3 dose escalation design with rituximab administered by intravenous infusion, 375 mg/m2 in four weekly doses, followed by a rest and four consolidation doses every 8 weeks for four cycles.

ALT-803 was administered concurrently at dose levels of 1 mcg/kg, 3 mcg/kg, and 6 mcg/kg IV followed by 6 mcg/kg, 10 mcg/kg, 15 mcg/kg, and 20 mcg/kg subcutaneously.

 

 


In total, 21 patients were treated: 16 patients had follicular lymphoma, four patients had marginal zone lymphoma, and one patient had small lymphocytic lymphoma. The median prior therapies received was two (range: 1-18) and five patients were treated who were refractory to prior anti-CD20 MAb therapy.

ALT-803 was well tolerated with no dose limiting toxicities or grade 4 or 5 adverse events. No patients discontinued ALT-803 and the recommended phase 2 dose was 20 mcg/kg subcutaneously. Grade 3 adverse events, regardless of attribution to ALT-803, included transient hypertension (14%), anemia (5%), nausea (5%), chills (5%), fever (5%), neutropenia (5%), and hyperglycemia (5%).

“Patients who received [subcutaneous] ALT-803 developed a unique injection site rash reaction that peaked 7-10 days later but resolved typically within 14 days. It was self-limited and resolved on its own,” Dr. Fehniger said.

At the time of the presentation, the best overall response rate was achieved in 11 of 21 patients (52%), with 9 complete responders (43%), and 2 partial responders (10%).
 

 


Of the 12 patients treated with ALT-803 subcutaneously, 11 patients had either stable disease, or partial or complete responses. All 11 patients remained on study and were in consolidation or follow-up and have not relapsed, Dr. Fehniger reported.

Among the five rituximab-refractory patients, the researchers observed one complete response, two patients with stable disease (45% and 36% tumor volume decrease), and two patients with partial disease. The durability of the responses can only be understood with longer follow-up, Dr. Fehniger said.

The peripheral blood of the patients was analyzed via flow cytometry and mass cytometry. Over the duration of four weekly doses, there was an increase in percentage (sixfold, P less than .001) and absolute number (10-fold, P less than .001) of natural killer cells at the 15-mcg/kg and 20-mcg/kg subcutaneous dose levels of ALT-803.

These results suggest that further studies of ALT-803 with other therapeutic targeting mAbs, or other immunotherapy modalities, are warranted, the researchers concluded.

Dr. Fehniger reported research funding from Altor BioScience.

SOURCE: Fehniger TA et al. AACR Annual Meeting, Abstract CT146.

 

– A combination of an immunostimulatory IL-15-based agent, ALT-803, with a therapeutic monoclonal antibody (mAb) against CD20, was well tolerated and had clinical activity in patients with indolent non-Hodgkin lymphoma (iNHL), according to preliminary findings from a phase 1 study.

“The cancer immunotherapy breakthrough that happened several years ago continues year after year, with a plethora of different modalities of immunotherapy at our disposal,” Todd A. Fehniger, MD, PhD, said at the annual meeting of the American Association for Cancer Research.

Immunotherapy with anti-CD20 mAbs, alone or in combination with chemotherapy, is a standard therapy for iNHL patients. Since iNHL cells express CD20, targeting it with mAbs triggers antitumor responses via cell surface receptors resulting in a potent antibody-dependent cellular toxicity. However, response in patients is highly heterogeneous, with relapse within a few months in a subset of patients. In addition, chemotherapeutic combinations can be toxic and result in serious and long-term complications.

“Relapsed or refractory iNHL is not curable and treatment strategies without long-term complications are needed,” said Dr. Fehniger, associate professor of medicine at Washington University, St. Louis.

In an attempt to address this, Dr. Fehniger and his colleagues combined rituximab, an anti-CD20 antibody, with a relatively new IL-15 agonist immunostimulatory agent called ALT-803.

In the phase 1 trial, the researchers enrolled patients with indolent non-Hodgkin lymphoma who had relapsed after at least 1 prior to CD20 antibody containing therapy. The study was a standard 3+3 dose escalation design with rituximab administered by intravenous infusion, 375 mg/m2 in four weekly doses, followed by a rest and four consolidation doses every 8 weeks for four cycles.

ALT-803 was administered concurrently at dose levels of 1 mcg/kg, 3 mcg/kg, and 6 mcg/kg IV followed by 6 mcg/kg, 10 mcg/kg, 15 mcg/kg, and 20 mcg/kg subcutaneously.

 

 


In total, 21 patients were treated: 16 patients had follicular lymphoma, four patients had marginal zone lymphoma, and one patient had small lymphocytic lymphoma. The median prior therapies received was two (range: 1-18) and five patients were treated who were refractory to prior anti-CD20 MAb therapy.

ALT-803 was well tolerated with no dose limiting toxicities or grade 4 or 5 adverse events. No patients discontinued ALT-803 and the recommended phase 2 dose was 20 mcg/kg subcutaneously. Grade 3 adverse events, regardless of attribution to ALT-803, included transient hypertension (14%), anemia (5%), nausea (5%), chills (5%), fever (5%), neutropenia (5%), and hyperglycemia (5%).

“Patients who received [subcutaneous] ALT-803 developed a unique injection site rash reaction that peaked 7-10 days later but resolved typically within 14 days. It was self-limited and resolved on its own,” Dr. Fehniger said.

At the time of the presentation, the best overall response rate was achieved in 11 of 21 patients (52%), with 9 complete responders (43%), and 2 partial responders (10%).
 

 


Of the 12 patients treated with ALT-803 subcutaneously, 11 patients had either stable disease, or partial or complete responses. All 11 patients remained on study and were in consolidation or follow-up and have not relapsed, Dr. Fehniger reported.

Among the five rituximab-refractory patients, the researchers observed one complete response, two patients with stable disease (45% and 36% tumor volume decrease), and two patients with partial disease. The durability of the responses can only be understood with longer follow-up, Dr. Fehniger said.

The peripheral blood of the patients was analyzed via flow cytometry and mass cytometry. Over the duration of four weekly doses, there was an increase in percentage (sixfold, P less than .001) and absolute number (10-fold, P less than .001) of natural killer cells at the 15-mcg/kg and 20-mcg/kg subcutaneous dose levels of ALT-803.

These results suggest that further studies of ALT-803 with other therapeutic targeting mAbs, or other immunotherapy modalities, are warranted, the researchers concluded.

Dr. Fehniger reported research funding from Altor BioScience.

SOURCE: Fehniger TA et al. AACR Annual Meeting, Abstract CT146.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

REPORTING FROM THE AACR ANNUAL MEETING

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: An IL-15 receptor super agonist plus rituximab was well tolerated in patients with indolent non-Hodgkin lymphoma.

Major finding: The ALT-803 plus rituximab combination achieved an overall response rate in 52% of patients, a complete response in 43%, and partial response in 10%.

Study details: A phase 1 study of 21 patients with indolent non-Hodgkin lymphoma.

Disclosures: Dr. Fehniger reported research funding from Altor BioScience LLC.

Source: Fehniger TA et al. AACR Annual Meeting, Abstract CT146.

Disqus Comments
Default
Use ProPublica

Early results favorable for combo TLR9 agonist + pembro in advanced melanoma

Article Type
Changed
Mon, 01/14/2019 - 10:21

 

– The intratumoral Toll-Like Receptor 9 (TLR-9) agonist, CMP-001, in combination with pembrolizumab in advanced melanoma patients, was well tolerated with a durable systemic clinical response, according to early results from an ongoing phase 1 trial.

Objective response rates on weekly (n = 56) and every 3 weeks schedules (n = 13) were 23% (13%-36%) and 15% (2%-45%) respectively, reported Mohammed M. Milhem, MBBS, of the University of Iowa, Iowa City.

For those dosed weekly at low dose (less than 5 mL) and high dose (5 mL or more), the ORR was 19% (n = 43, 95% confidence interval, 8%-33%) and 27% (n = 26, 95% CI, 12%-48%), respectively. Activity was demonstrated in subjects regardless of tumor burden, Dr. Milhem said at the annual meeting of the American Association for Cancer Research.

In this phase 1b study with a 3+3 design of dose escalation and expansion, the researchers enrolled patients with advanced melanoma who did not respond or had progressed resistant on prior anti-PD-1 monotherapy or in combination. CMP-001 was injected intratumorally in combination with pembrolizumab as per label intravenously.

The study drug CMP-001 has two components, a 30-mer CpG-A DNA oligonucleotide and a nonvirulent virus-like particle (VLP). The CpG-A DNA is packaged within the VLP that protects it from degradation and also allows TLR9 receptor uptake. CpG-A DNA acts as a TLR9 agonist by binding to it, thereby activating plasmacytoid dendritic cells (pDCs) within the tumor microenvironment. The activation results in secretion of large amounts of type 1 interferon and Th1 chemokines, changing the microenvironment from a “cold/desert-like” immune suppressed state to a “hot” antitumor inflamed state, Dr. Milhem said.

“The T cells thus generated can mediate tumor rejection both in the injected and noninjected tumor,” he said. Two CMP-001 schedules were evaluated, weekly for 7 weeks or weekly for 2 weeks, followed thereafter by every 3 weeks until discontinuation (due to progression, toxicity, investigator decision, or withdrawal of consent). Scans were done every 12 weeks and tumor response was assessed by RECIST v1.1.

The CMP-001 dose escalation scheme ranged from 1 mg to 10 mg. The maximum tolerated dose was not reached and the dose of 5 mg/weekly plus pembrolizumab was used for the dose expansion phase. It was up to the investigator to increase the dose to 10 mg since maximum tolerated dose was not reached. The key inclusion criteria were metastatic or unresectable melanoma; in the dose escalation phase prior best response to anti-PD1-based therapy was disease progression or stable disease. In the dose expansion phase, patients who had progressed on anti-PD1 based therapy were allowed regardless of best response. There was no restriction on the number of prior lines of therapy.

 

 


A total of 69 subjects were treated, 44 subjects from dose escalation and 25 in the expansion phase (ongoing). Two subjects discontinued because of treatment-related adverse events. The rest of the patients had a manageable toxicity profile consisting predominantly of fever, nausea/vomiting, hypotension and rigors. Severe grade 3/4 treatment-related adverse events were reported in more than 1 subject, with hypotension (n = 9, 13%) being the most prominent AE, followed by anemia (n = 2, 3%), chills (n = 2, 3%), and hypertension (n = 2, 3%). Hypotension was manageable by responsive fluid resuscitation and in some patients required stress dose steroids. Most of these side effects occurred 1-4 hours after the CMP-001 injection.

Of the 18 responders, 1 progressed, 2 withdrew consent, and 13 remain on study with 2 subjects maintaining their response though week 72. The median duration of response was not reached. Regression of noninjected tumors occurred in cutaneous, nodal, hepatic, and splenic metastases.

“CMP-001 plus pembrolizumab induced systemic antitumor activity, and not just local efficacy since both injected and noninjected target lesions changed from baseline per RECIST,” Dr. Milhem said. Not only did the responders show a rapid reduction in target lesions from baseline, but also a durable tumor regression as usually seen with other immunotherapeutics.

Immunohistochemical analysis of tumor biopsies demonstrated increase in CD8 (greater than fivefold) and PD-L1 expression, 5 weeks after therapy in a subset of patients with pre- and posttreatment biopsies. Transcriptional analysis by RNA-seq revealed induction of T cell inflamed gene signature, notably significant upregulation of TLR, and IFN-responsive genes.

 

 


It would be interesting to further investigate how this combination therapy compares with other strategies in a similar clinical scenario, such as oncolytic virus, other TLR ligands or means of APC activation, discussant Jedd Wolchok, MD, PhD, pointed out. Understanding resistance mechanisms at an individual patient level and optimal patient selection for this combination therapy remains a challenge, he said.

Dr. Milhem had no financial relationships to disclose.

SOURCE: Milhem MD et al. AACR Annual Meeting Abstract CT144.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– The intratumoral Toll-Like Receptor 9 (TLR-9) agonist, CMP-001, in combination with pembrolizumab in advanced melanoma patients, was well tolerated with a durable systemic clinical response, according to early results from an ongoing phase 1 trial.

Objective response rates on weekly (n = 56) and every 3 weeks schedules (n = 13) were 23% (13%-36%) and 15% (2%-45%) respectively, reported Mohammed M. Milhem, MBBS, of the University of Iowa, Iowa City.

For those dosed weekly at low dose (less than 5 mL) and high dose (5 mL or more), the ORR was 19% (n = 43, 95% confidence interval, 8%-33%) and 27% (n = 26, 95% CI, 12%-48%), respectively. Activity was demonstrated in subjects regardless of tumor burden, Dr. Milhem said at the annual meeting of the American Association for Cancer Research.

In this phase 1b study with a 3+3 design of dose escalation and expansion, the researchers enrolled patients with advanced melanoma who did not respond or had progressed resistant on prior anti-PD-1 monotherapy or in combination. CMP-001 was injected intratumorally in combination with pembrolizumab as per label intravenously.

The study drug CMP-001 has two components, a 30-mer CpG-A DNA oligonucleotide and a nonvirulent virus-like particle (VLP). The CpG-A DNA is packaged within the VLP that protects it from degradation and also allows TLR9 receptor uptake. CpG-A DNA acts as a TLR9 agonist by binding to it, thereby activating plasmacytoid dendritic cells (pDCs) within the tumor microenvironment. The activation results in secretion of large amounts of type 1 interferon and Th1 chemokines, changing the microenvironment from a “cold/desert-like” immune suppressed state to a “hot” antitumor inflamed state, Dr. Milhem said.

“The T cells thus generated can mediate tumor rejection both in the injected and noninjected tumor,” he said. Two CMP-001 schedules were evaluated, weekly for 7 weeks or weekly for 2 weeks, followed thereafter by every 3 weeks until discontinuation (due to progression, toxicity, investigator decision, or withdrawal of consent). Scans were done every 12 weeks and tumor response was assessed by RECIST v1.1.

The CMP-001 dose escalation scheme ranged from 1 mg to 10 mg. The maximum tolerated dose was not reached and the dose of 5 mg/weekly plus pembrolizumab was used for the dose expansion phase. It was up to the investigator to increase the dose to 10 mg since maximum tolerated dose was not reached. The key inclusion criteria were metastatic or unresectable melanoma; in the dose escalation phase prior best response to anti-PD1-based therapy was disease progression or stable disease. In the dose expansion phase, patients who had progressed on anti-PD1 based therapy were allowed regardless of best response. There was no restriction on the number of prior lines of therapy.

 

 


A total of 69 subjects were treated, 44 subjects from dose escalation and 25 in the expansion phase (ongoing). Two subjects discontinued because of treatment-related adverse events. The rest of the patients had a manageable toxicity profile consisting predominantly of fever, nausea/vomiting, hypotension and rigors. Severe grade 3/4 treatment-related adverse events were reported in more than 1 subject, with hypotension (n = 9, 13%) being the most prominent AE, followed by anemia (n = 2, 3%), chills (n = 2, 3%), and hypertension (n = 2, 3%). Hypotension was manageable by responsive fluid resuscitation and in some patients required stress dose steroids. Most of these side effects occurred 1-4 hours after the CMP-001 injection.

Of the 18 responders, 1 progressed, 2 withdrew consent, and 13 remain on study with 2 subjects maintaining their response though week 72. The median duration of response was not reached. Regression of noninjected tumors occurred in cutaneous, nodal, hepatic, and splenic metastases.

“CMP-001 plus pembrolizumab induced systemic antitumor activity, and not just local efficacy since both injected and noninjected target lesions changed from baseline per RECIST,” Dr. Milhem said. Not only did the responders show a rapid reduction in target lesions from baseline, but also a durable tumor regression as usually seen with other immunotherapeutics.

Immunohistochemical analysis of tumor biopsies demonstrated increase in CD8 (greater than fivefold) and PD-L1 expression, 5 weeks after therapy in a subset of patients with pre- and posttreatment biopsies. Transcriptional analysis by RNA-seq revealed induction of T cell inflamed gene signature, notably significant upregulation of TLR, and IFN-responsive genes.

 

 


It would be interesting to further investigate how this combination therapy compares with other strategies in a similar clinical scenario, such as oncolytic virus, other TLR ligands or means of APC activation, discussant Jedd Wolchok, MD, PhD, pointed out. Understanding resistance mechanisms at an individual patient level and optimal patient selection for this combination therapy remains a challenge, he said.

Dr. Milhem had no financial relationships to disclose.

SOURCE: Milhem MD et al. AACR Annual Meeting Abstract CT144.

 

– The intratumoral Toll-Like Receptor 9 (TLR-9) agonist, CMP-001, in combination with pembrolizumab in advanced melanoma patients, was well tolerated with a durable systemic clinical response, according to early results from an ongoing phase 1 trial.

Objective response rates on weekly (n = 56) and every 3 weeks schedules (n = 13) were 23% (13%-36%) and 15% (2%-45%) respectively, reported Mohammed M. Milhem, MBBS, of the University of Iowa, Iowa City.

For those dosed weekly at low dose (less than 5 mL) and high dose (5 mL or more), the ORR was 19% (n = 43, 95% confidence interval, 8%-33%) and 27% (n = 26, 95% CI, 12%-48%), respectively. Activity was demonstrated in subjects regardless of tumor burden, Dr. Milhem said at the annual meeting of the American Association for Cancer Research.

In this phase 1b study with a 3+3 design of dose escalation and expansion, the researchers enrolled patients with advanced melanoma who did not respond or had progressed resistant on prior anti-PD-1 monotherapy or in combination. CMP-001 was injected intratumorally in combination with pembrolizumab as per label intravenously.

The study drug CMP-001 has two components, a 30-mer CpG-A DNA oligonucleotide and a nonvirulent virus-like particle (VLP). The CpG-A DNA is packaged within the VLP that protects it from degradation and also allows TLR9 receptor uptake. CpG-A DNA acts as a TLR9 agonist by binding to it, thereby activating plasmacytoid dendritic cells (pDCs) within the tumor microenvironment. The activation results in secretion of large amounts of type 1 interferon and Th1 chemokines, changing the microenvironment from a “cold/desert-like” immune suppressed state to a “hot” antitumor inflamed state, Dr. Milhem said.

“The T cells thus generated can mediate tumor rejection both in the injected and noninjected tumor,” he said. Two CMP-001 schedules were evaluated, weekly for 7 weeks or weekly for 2 weeks, followed thereafter by every 3 weeks until discontinuation (due to progression, toxicity, investigator decision, or withdrawal of consent). Scans were done every 12 weeks and tumor response was assessed by RECIST v1.1.

The CMP-001 dose escalation scheme ranged from 1 mg to 10 mg. The maximum tolerated dose was not reached and the dose of 5 mg/weekly plus pembrolizumab was used for the dose expansion phase. It was up to the investigator to increase the dose to 10 mg since maximum tolerated dose was not reached. The key inclusion criteria were metastatic or unresectable melanoma; in the dose escalation phase prior best response to anti-PD1-based therapy was disease progression or stable disease. In the dose expansion phase, patients who had progressed on anti-PD1 based therapy were allowed regardless of best response. There was no restriction on the number of prior lines of therapy.

 

 


A total of 69 subjects were treated, 44 subjects from dose escalation and 25 in the expansion phase (ongoing). Two subjects discontinued because of treatment-related adverse events. The rest of the patients had a manageable toxicity profile consisting predominantly of fever, nausea/vomiting, hypotension and rigors. Severe grade 3/4 treatment-related adverse events were reported in more than 1 subject, with hypotension (n = 9, 13%) being the most prominent AE, followed by anemia (n = 2, 3%), chills (n = 2, 3%), and hypertension (n = 2, 3%). Hypotension was manageable by responsive fluid resuscitation and in some patients required stress dose steroids. Most of these side effects occurred 1-4 hours after the CMP-001 injection.

Of the 18 responders, 1 progressed, 2 withdrew consent, and 13 remain on study with 2 subjects maintaining their response though week 72. The median duration of response was not reached. Regression of noninjected tumors occurred in cutaneous, nodal, hepatic, and splenic metastases.

“CMP-001 plus pembrolizumab induced systemic antitumor activity, and not just local efficacy since both injected and noninjected target lesions changed from baseline per RECIST,” Dr. Milhem said. Not only did the responders show a rapid reduction in target lesions from baseline, but also a durable tumor regression as usually seen with other immunotherapeutics.

Immunohistochemical analysis of tumor biopsies demonstrated increase in CD8 (greater than fivefold) and PD-L1 expression, 5 weeks after therapy in a subset of patients with pre- and posttreatment biopsies. Transcriptional analysis by RNA-seq revealed induction of T cell inflamed gene signature, notably significant upregulation of TLR, and IFN-responsive genes.

 

 


It would be interesting to further investigate how this combination therapy compares with other strategies in a similar clinical scenario, such as oncolytic virus, other TLR ligands or means of APC activation, discussant Jedd Wolchok, MD, PhD, pointed out. Understanding resistance mechanisms at an individual patient level and optimal patient selection for this combination therapy remains a challenge, he said.

Dr. Milhem had no financial relationships to disclose.

SOURCE: Milhem MD et al. AACR Annual Meeting Abstract CT144.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

REPORTING FROM THE AACR ANNUAL MEETING

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: The combination demonstrated a manageable toxicity profile with ORR of 22%.

Major finding: Objective response rates on weekly (n = 56) and every 3 weeks schedules (n = 13) were 23% (13%-36%) and 15% (2%-45%) respectively.

Study details: This phase 1b study comprised 69 patients (44 in escalation and 25 in expansion).

Disclosures: Dr. Milhem had no financial relationships to disclose.

Source: Milhem MD et al. AACR Annual Meeting. Abstract CT144.

Disqus Comments
Default
Use ProPublica

Nivolumab plus ipilimumab boosts PFS in advanced NSCLC with high tumor mutational burden

Article Type
Changed
Fri, 01/04/2019 - 14:16

 

– The combination of nivolumab plus ipilimumab prolonged progression-free survival (PFS), in comparison with platinum based chemotherapy, as initial treatment for advanced non–small cell lung cancer (NSCLC) patients with a high tumor mutation burden (TMB), regardless of PDL-1 status, according to the first analysis of Checkmate 227.

Median PFS was 7.2 months for patients receiving the immunotherapy combination (95% confidence interval, 5.5-13.2) versus 5.5 months (95% CI, 4.4-5.8) for those receiving platinum-based chemotherapy (hazard ratio for disease progression or death, 0.58; 97.5% CI, 0.41-0.81; P less than .001).

The trial results validate the benefit of nivolumab plus ipilimumab in NSCLC and the role of tumor mutational burden as a biomarker for patient selection, Matthew D. Hellmann, MD, said at the annual meeting of the American Association for Cancer Research.

“Checkmate 227 is the pivotal phase 3 study to validate TMB as an important and independent biomarker to be routinely tested in treatment-naive advanced NSCLC,” he said.

Overall survival data with the combination versus chemotherapy was “encouraging” but not yet complete in patients with high TMB, Dr. Hellmann said.

Results from this first analysis (part 1) of Checkmate 227, a multipart trial, were simultaneously published in the New England Journal of Medicine.

The trial was designed prior to emerging data about TMB. The study was later amended prior to initial analysis to include a second coprimary endpoint evaluating PFS with nivolumab plus ipilimumab versus chemotherapy among patients with a tumor mutational burden of at least 10 mutations per megabase (mut/Mb), irrespective of PD-L1 expression level.

 

 


For part 1 of the trial, patients were randomized 1:1:1 to nivolumab 3 mg/kg every 2 weeks plus low-dose ipilimumab 1 mg/kg every 6 weeks; histology-based platinum-doublet chemotherapy every 3 weeks for up to four cycles; and nivolumab 240 mg every 2 weeks or nivolumab 360 mg plus histology-based platinum-doublet chemotherapy every 3 weeks for up to four cycles, followed by nivolumab monotherapy.

Of all randomized patients in part 1 (n=1,739), 1,004 (58%) were evaluable for TMB analyses. Of all TMB-evaluable patients, 444 (44%) had TMB greater than or equal to 10 mut/Mb, including 139 patients randomized to nivolumab plus ipilimumab and 160 patients randomized to chemotherapy. TMB was assessed using the validated assay, FoundationOne CDx.

One-year PFS was higher with nivolumab plus ipilimumab than with chemotherapy among all randomized patients in part 1 (30.9% vs. 17.0%; HR for disease progression or death, 0.83; 95% CI, 0.72-0.96).

The PFS was significantly prolonged with the combination, compared with chemotherapy among patients with a high TMB; the 1-year PFS rate was 42.6% versus 13.2%, respectively, and the median PFS was 7.2 months (95% CI, 5.5-13.2) versus 5.5 months (95% CI, 4.4-5.8) respectively (HR for disease progression or death, 0.58; 97.5% CI, 0.41-0.81; P less than .001). In a subgroup analysis, PFS was longer with nivolumab plus ipilimumab versus chemotherapy for patients with a programmed death ligand 1 expression level of at least 1% and those with a level of less than 1%, reported Dr. Hellmann of Memorial Sloan Kettering Cancer Center, New York.

 

 


Safety was manageable and consistent with previous reports of nivolumab plus low-dose ipilimumab in NSCLC. Grade 3-4 treatment-related adverse events with the combination were skin reactions (34%), endocrine (23%), gastrointestinal (18%), hepatic (15%), pulmonary (8%), hypersensitivity (4%), and renal (4%) events. Overall, treatment-related deaths occurred in 1% of patients treated in both the combination and chemotherapy arms.

Results from Checkmate 227 may introduce two new standards of care for the first-line treatment of NSCLC, Dr. Hellmann said. The immunotherapy combination is introduced as a new option for the first-line treatment of NSCLC with a high TMB, sparing first-line chemotherapy, and it validates TMB as an “important and independent biomarker to be routinely tested in treatment-naive, advanced NSCLC,” he concluded.

Dr. Hellmann disclosed relationships with Genentech, Bristol-Myers Squibb, Merck, AstraZeneca, Novartis, Janssen, Mirati, and Shattuck Labs.

SOURCE: Hellmann MD et al. AACR Annual Meeting, Abstract CT077.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– The combination of nivolumab plus ipilimumab prolonged progression-free survival (PFS), in comparison with platinum based chemotherapy, as initial treatment for advanced non–small cell lung cancer (NSCLC) patients with a high tumor mutation burden (TMB), regardless of PDL-1 status, according to the first analysis of Checkmate 227.

Median PFS was 7.2 months for patients receiving the immunotherapy combination (95% confidence interval, 5.5-13.2) versus 5.5 months (95% CI, 4.4-5.8) for those receiving platinum-based chemotherapy (hazard ratio for disease progression or death, 0.58; 97.5% CI, 0.41-0.81; P less than .001).

The trial results validate the benefit of nivolumab plus ipilimumab in NSCLC and the role of tumor mutational burden as a biomarker for patient selection, Matthew D. Hellmann, MD, said at the annual meeting of the American Association for Cancer Research.

“Checkmate 227 is the pivotal phase 3 study to validate TMB as an important and independent biomarker to be routinely tested in treatment-naive advanced NSCLC,” he said.

Overall survival data with the combination versus chemotherapy was “encouraging” but not yet complete in patients with high TMB, Dr. Hellmann said.

Results from this first analysis (part 1) of Checkmate 227, a multipart trial, were simultaneously published in the New England Journal of Medicine.

The trial was designed prior to emerging data about TMB. The study was later amended prior to initial analysis to include a second coprimary endpoint evaluating PFS with nivolumab plus ipilimumab versus chemotherapy among patients with a tumor mutational burden of at least 10 mutations per megabase (mut/Mb), irrespective of PD-L1 expression level.

 

 


For part 1 of the trial, patients were randomized 1:1:1 to nivolumab 3 mg/kg every 2 weeks plus low-dose ipilimumab 1 mg/kg every 6 weeks; histology-based platinum-doublet chemotherapy every 3 weeks for up to four cycles; and nivolumab 240 mg every 2 weeks or nivolumab 360 mg plus histology-based platinum-doublet chemotherapy every 3 weeks for up to four cycles, followed by nivolumab monotherapy.

Of all randomized patients in part 1 (n=1,739), 1,004 (58%) were evaluable for TMB analyses. Of all TMB-evaluable patients, 444 (44%) had TMB greater than or equal to 10 mut/Mb, including 139 patients randomized to nivolumab plus ipilimumab and 160 patients randomized to chemotherapy. TMB was assessed using the validated assay, FoundationOne CDx.

One-year PFS was higher with nivolumab plus ipilimumab than with chemotherapy among all randomized patients in part 1 (30.9% vs. 17.0%; HR for disease progression or death, 0.83; 95% CI, 0.72-0.96).

The PFS was significantly prolonged with the combination, compared with chemotherapy among patients with a high TMB; the 1-year PFS rate was 42.6% versus 13.2%, respectively, and the median PFS was 7.2 months (95% CI, 5.5-13.2) versus 5.5 months (95% CI, 4.4-5.8) respectively (HR for disease progression or death, 0.58; 97.5% CI, 0.41-0.81; P less than .001). In a subgroup analysis, PFS was longer with nivolumab plus ipilimumab versus chemotherapy for patients with a programmed death ligand 1 expression level of at least 1% and those with a level of less than 1%, reported Dr. Hellmann of Memorial Sloan Kettering Cancer Center, New York.

 

 


Safety was manageable and consistent with previous reports of nivolumab plus low-dose ipilimumab in NSCLC. Grade 3-4 treatment-related adverse events with the combination were skin reactions (34%), endocrine (23%), gastrointestinal (18%), hepatic (15%), pulmonary (8%), hypersensitivity (4%), and renal (4%) events. Overall, treatment-related deaths occurred in 1% of patients treated in both the combination and chemotherapy arms.

Results from Checkmate 227 may introduce two new standards of care for the first-line treatment of NSCLC, Dr. Hellmann said. The immunotherapy combination is introduced as a new option for the first-line treatment of NSCLC with a high TMB, sparing first-line chemotherapy, and it validates TMB as an “important and independent biomarker to be routinely tested in treatment-naive, advanced NSCLC,” he concluded.

Dr. Hellmann disclosed relationships with Genentech, Bristol-Myers Squibb, Merck, AstraZeneca, Novartis, Janssen, Mirati, and Shattuck Labs.

SOURCE: Hellmann MD et al. AACR Annual Meeting, Abstract CT077.

 

– The combination of nivolumab plus ipilimumab prolonged progression-free survival (PFS), in comparison with platinum based chemotherapy, as initial treatment for advanced non–small cell lung cancer (NSCLC) patients with a high tumor mutation burden (TMB), regardless of PDL-1 status, according to the first analysis of Checkmate 227.

Median PFS was 7.2 months for patients receiving the immunotherapy combination (95% confidence interval, 5.5-13.2) versus 5.5 months (95% CI, 4.4-5.8) for those receiving platinum-based chemotherapy (hazard ratio for disease progression or death, 0.58; 97.5% CI, 0.41-0.81; P less than .001).

The trial results validate the benefit of nivolumab plus ipilimumab in NSCLC and the role of tumor mutational burden as a biomarker for patient selection, Matthew D. Hellmann, MD, said at the annual meeting of the American Association for Cancer Research.

“Checkmate 227 is the pivotal phase 3 study to validate TMB as an important and independent biomarker to be routinely tested in treatment-naive advanced NSCLC,” he said.

Overall survival data with the combination versus chemotherapy was “encouraging” but not yet complete in patients with high TMB, Dr. Hellmann said.

Results from this first analysis (part 1) of Checkmate 227, a multipart trial, were simultaneously published in the New England Journal of Medicine.

The trial was designed prior to emerging data about TMB. The study was later amended prior to initial analysis to include a second coprimary endpoint evaluating PFS with nivolumab plus ipilimumab versus chemotherapy among patients with a tumor mutational burden of at least 10 mutations per megabase (mut/Mb), irrespective of PD-L1 expression level.

 

 


For part 1 of the trial, patients were randomized 1:1:1 to nivolumab 3 mg/kg every 2 weeks plus low-dose ipilimumab 1 mg/kg every 6 weeks; histology-based platinum-doublet chemotherapy every 3 weeks for up to four cycles; and nivolumab 240 mg every 2 weeks or nivolumab 360 mg plus histology-based platinum-doublet chemotherapy every 3 weeks for up to four cycles, followed by nivolumab monotherapy.

Of all randomized patients in part 1 (n=1,739), 1,004 (58%) were evaluable for TMB analyses. Of all TMB-evaluable patients, 444 (44%) had TMB greater than or equal to 10 mut/Mb, including 139 patients randomized to nivolumab plus ipilimumab and 160 patients randomized to chemotherapy. TMB was assessed using the validated assay, FoundationOne CDx.

One-year PFS was higher with nivolumab plus ipilimumab than with chemotherapy among all randomized patients in part 1 (30.9% vs. 17.0%; HR for disease progression or death, 0.83; 95% CI, 0.72-0.96).

The PFS was significantly prolonged with the combination, compared with chemotherapy among patients with a high TMB; the 1-year PFS rate was 42.6% versus 13.2%, respectively, and the median PFS was 7.2 months (95% CI, 5.5-13.2) versus 5.5 months (95% CI, 4.4-5.8) respectively (HR for disease progression or death, 0.58; 97.5% CI, 0.41-0.81; P less than .001). In a subgroup analysis, PFS was longer with nivolumab plus ipilimumab versus chemotherapy for patients with a programmed death ligand 1 expression level of at least 1% and those with a level of less than 1%, reported Dr. Hellmann of Memorial Sloan Kettering Cancer Center, New York.

 

 


Safety was manageable and consistent with previous reports of nivolumab plus low-dose ipilimumab in NSCLC. Grade 3-4 treatment-related adverse events with the combination were skin reactions (34%), endocrine (23%), gastrointestinal (18%), hepatic (15%), pulmonary (8%), hypersensitivity (4%), and renal (4%) events. Overall, treatment-related deaths occurred in 1% of patients treated in both the combination and chemotherapy arms.

Results from Checkmate 227 may introduce two new standards of care for the first-line treatment of NSCLC, Dr. Hellmann said. The immunotherapy combination is introduced as a new option for the first-line treatment of NSCLC with a high TMB, sparing first-line chemotherapy, and it validates TMB as an “important and independent biomarker to be routinely tested in treatment-naive, advanced NSCLC,” he concluded.

Dr. Hellmann disclosed relationships with Genentech, Bristol-Myers Squibb, Merck, AstraZeneca, Novartis, Janssen, Mirati, and Shattuck Labs.

SOURCE: Hellmann MD et al. AACR Annual Meeting, Abstract CT077.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

REPORTING FROM THE AACR ANNUAL MEETING

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: PFS was prolonged with the immunotherapy combination of nivolumab plus low-dose ipilimumab versus chemotherapy in first-line advanced NSCLC patients with high tumor mutational burden, independent of PD-L1 expression or tumor histology.

Major finding: Median progression-free survival was 7.2 months (95% CI, 5.5-13.2) for the immunotherapy combination versus 5.5 months (95% CI, 4.4-5.8) for chemotherapy (HR, 0.58; 97.5% CI, 0.41-0.81; P less than .001).

Study details: CheckMate-227 is a multipart open-label phase 3 trial evaluating nivolumab-based combinations versus platinum-doublet chemotherapy in patients with first-line advanced non–small cell lung cancer across nonsquamous and squamous tumor histology.

Disclosures: Dr. Hellmann disclosed relationships with Genentech, Bristol-Myers Squibb, Merck, AstraZeneca, Novartis, Janssen, Mirati, and Shattuck Labs.

Source: Hellmann MD et al. AACR Annual Meeting, Abstract CT077.

Disqus Comments
Default
Use ProPublica

OlympiAD: No statistically significant boost in OS with olaparib in HER2-negative mBC

Article Type
Changed
Wed, 01/04/2023 - 16:45

 

– Median overall survival (OS) in patients with HER2-negative metastatic breast cancer (mBC) and germline BRCA mutation (gBRCAm), although not statistically significant, was 2.2 months longer with olaparib versus physician’s choice chemotherapy (TPC), according to the final analysis of the OlympiAD study.

The results suggested the possibility of greater benefit among chemotherapy naive patients for metastatic breast cancer, with no cumulative toxicity reported with extended exposure, Mark E. Robson, MD, said at the annual meeting of the American Association for Cancer Research.

OlympiAD was a randomized, controlled, open-label, multicenter, phase 3 study of olaparib tablet monotherapy (300 mg, twice daily) compared with predeclared TPC monotherapy (capecitabine, vinorelbine, or eribulin). Patients were stratified by prior chemotherapy, prior platinum, and receptor status (ER+ and/or PR+ vs. TNBC). Of 302 randomized patients, 205 received olaparib and 91 received TPC (6 TPC patients declined treatment). Eligible patients had HER2-negative mBC and a germline BRCA mutation. In addition, patients should have received less than or equal to two chemotherapy lines in the metastatic setting, with prior anthracycline and taxane treatment either as (neo)adjuvant therapy or in the metastatic setting.

The data presented at AACR was a follow-up on the primary progression-free survival (PFS) analysis, which demonstrated significant benefit in olaparib over standard chemotherapy TPC (7.0 vs 4.2 months, HR 0.58, 95% confidence interval, 0.43-0.80, P less than .001). Overall response rate (ORR) in the olaparib arm was double of that observed on the TPC arm in measurable disease patients (59.9% vs. 28.8%).

 

 


At the final OS analysis with 192 deaths, HR for OS in the olaparib vs TPC group was 0.90 (95% CI, 0.66-1.23; P = .513), reported Dr. Robson of Memorial Sloan Kettering Cancer Center, New York.

“The preplanned subgroup analyses according to the stratification factors were not powered to detect survival advantages, and were considered only hypothesis generating,” he said.

In patients who had not received chemotherapy in the metastatic setting, there was a median difference in OS of 7.9 months with olaparib (HR 0.51, 95% CI, 0.29-0.90; nominal P = .02; median 22.6 vs. 14.7 months).

Median follow-up for OS was 18.9 months for olaparib vs. 15.5 months in the TPC group.

No differences were observed between patients that were ER and/or PgR positive vs. TNBC, or whether patients received prior platinum, Dr. Robson said.

Grade 3 adverse events were similar to those in the primary analysis with no cumulative toxicity with extended exposure, he said.

SOURCE: Robson ME et al. AACR Annual Meeting Abstract CT038.

Meeting/Event
Publications
Topics
Sections
Meeting/Event
Meeting/Event

 

– Median overall survival (OS) in patients with HER2-negative metastatic breast cancer (mBC) and germline BRCA mutation (gBRCAm), although not statistically significant, was 2.2 months longer with olaparib versus physician’s choice chemotherapy (TPC), according to the final analysis of the OlympiAD study.

The results suggested the possibility of greater benefit among chemotherapy naive patients for metastatic breast cancer, with no cumulative toxicity reported with extended exposure, Mark E. Robson, MD, said at the annual meeting of the American Association for Cancer Research.

OlympiAD was a randomized, controlled, open-label, multicenter, phase 3 study of olaparib tablet monotherapy (300 mg, twice daily) compared with predeclared TPC monotherapy (capecitabine, vinorelbine, or eribulin). Patients were stratified by prior chemotherapy, prior platinum, and receptor status (ER+ and/or PR+ vs. TNBC). Of 302 randomized patients, 205 received olaparib and 91 received TPC (6 TPC patients declined treatment). Eligible patients had HER2-negative mBC and a germline BRCA mutation. In addition, patients should have received less than or equal to two chemotherapy lines in the metastatic setting, with prior anthracycline and taxane treatment either as (neo)adjuvant therapy or in the metastatic setting.

The data presented at AACR was a follow-up on the primary progression-free survival (PFS) analysis, which demonstrated significant benefit in olaparib over standard chemotherapy TPC (7.0 vs 4.2 months, HR 0.58, 95% confidence interval, 0.43-0.80, P less than .001). Overall response rate (ORR) in the olaparib arm was double of that observed on the TPC arm in measurable disease patients (59.9% vs. 28.8%).

 

 


At the final OS analysis with 192 deaths, HR for OS in the olaparib vs TPC group was 0.90 (95% CI, 0.66-1.23; P = .513), reported Dr. Robson of Memorial Sloan Kettering Cancer Center, New York.

“The preplanned subgroup analyses according to the stratification factors were not powered to detect survival advantages, and were considered only hypothesis generating,” he said.

In patients who had not received chemotherapy in the metastatic setting, there was a median difference in OS of 7.9 months with olaparib (HR 0.51, 95% CI, 0.29-0.90; nominal P = .02; median 22.6 vs. 14.7 months).

Median follow-up for OS was 18.9 months for olaparib vs. 15.5 months in the TPC group.

No differences were observed between patients that were ER and/or PgR positive vs. TNBC, or whether patients received prior platinum, Dr. Robson said.

Grade 3 adverse events were similar to those in the primary analysis with no cumulative toxicity with extended exposure, he said.

SOURCE: Robson ME et al. AACR Annual Meeting Abstract CT038.

 

– Median overall survival (OS) in patients with HER2-negative metastatic breast cancer (mBC) and germline BRCA mutation (gBRCAm), although not statistically significant, was 2.2 months longer with olaparib versus physician’s choice chemotherapy (TPC), according to the final analysis of the OlympiAD study.

The results suggested the possibility of greater benefit among chemotherapy naive patients for metastatic breast cancer, with no cumulative toxicity reported with extended exposure, Mark E. Robson, MD, said at the annual meeting of the American Association for Cancer Research.

OlympiAD was a randomized, controlled, open-label, multicenter, phase 3 study of olaparib tablet monotherapy (300 mg, twice daily) compared with predeclared TPC monotherapy (capecitabine, vinorelbine, or eribulin). Patients were stratified by prior chemotherapy, prior platinum, and receptor status (ER+ and/or PR+ vs. TNBC). Of 302 randomized patients, 205 received olaparib and 91 received TPC (6 TPC patients declined treatment). Eligible patients had HER2-negative mBC and a germline BRCA mutation. In addition, patients should have received less than or equal to two chemotherapy lines in the metastatic setting, with prior anthracycline and taxane treatment either as (neo)adjuvant therapy or in the metastatic setting.

The data presented at AACR was a follow-up on the primary progression-free survival (PFS) analysis, which demonstrated significant benefit in olaparib over standard chemotherapy TPC (7.0 vs 4.2 months, HR 0.58, 95% confidence interval, 0.43-0.80, P less than .001). Overall response rate (ORR) in the olaparib arm was double of that observed on the TPC arm in measurable disease patients (59.9% vs. 28.8%).

 

 


At the final OS analysis with 192 deaths, HR for OS in the olaparib vs TPC group was 0.90 (95% CI, 0.66-1.23; P = .513), reported Dr. Robson of Memorial Sloan Kettering Cancer Center, New York.

“The preplanned subgroup analyses according to the stratification factors were not powered to detect survival advantages, and were considered only hypothesis generating,” he said.

In patients who had not received chemotherapy in the metastatic setting, there was a median difference in OS of 7.9 months with olaparib (HR 0.51, 95% CI, 0.29-0.90; nominal P = .02; median 22.6 vs. 14.7 months).

Median follow-up for OS was 18.9 months for olaparib vs. 15.5 months in the TPC group.

No differences were observed between patients that were ER and/or PgR positive vs. TNBC, or whether patients received prior platinum, Dr. Robson said.

Grade 3 adverse events were similar to those in the primary analysis with no cumulative toxicity with extended exposure, he said.

SOURCE: Robson ME et al. AACR Annual Meeting Abstract CT038.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM THE AACR ANNUAL MEETING

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Median overall survival was not significantly different with olaparib versus chemotherapy in patients with BRCA-mutated, HER2-negative metastatic breast cancer.

Major finding: Median overall survival in patients with HER2-negative metastatic breast cancer and a germline BRCA mutation was 19.3 months versus 17.1 months for olaparib versus chemotherapy (HR 0.90 95% CI 0.66, 1.23; P = .513).

Study details: Randomized, controlled, open-label, phase 3 trial (OlympiAD) of olaparib tablet monotherapy (300 mg, twice daily) compared with predeclared physician’s choice chemotherapy (capecitabine, vinorelbine, or eribulin).

Disclosures: Dr. Robson disclosed relationships with AstraZeneca, AbbVie, McKesson, Myriad Genetics, and Medivation.

Source: Robson ME et al. AACR Annual Meeting Abstract CT038.

Disqus Comments
Default
Use ProPublica

AGA Clinical Practice Update: Incorporating psychological care in the management of chronic digestive diseases

Article Type
Changed
Fri, 01/18/2019 - 17:28

Psychogastroenterology is the science of applying psychological principles and techniques to alleviate the burden of chronic digestive diseases. This burden includes digestive symptoms and disease severity, as well as patients’ ability to cope with them. Chronic digestive diseases, such as irritable bowel syndrome, gastroesophageal reflux disease, and inflammatory bowel diseases, cannot be disentangled from their psychosocial context. In this regard, the role of gastroenterologists in promoting best practices for the assessment and referral of patients across the spectrum of disease to brain-gut psychotherapies is crucial.

A review by Laurie Keefer, PhD, AGAF, and her coauthors, published in the April issue of Gastroenterology, provided a clinical update on the structure and efficacy of two major classes of psychogastroenterology – cognitive-behavioral therapy (CBT) and gut-directed hypnotherapy (HYP). The review discussed the effects of these therapies on GI symptoms and the patients’ ability to improve coping, resilience, and self-regulation. The review also provided a framework to understand the scientific rationale and best practices associated with incorporating brain-gut psychotherapies into routine GI care. Furthermore, it presented recommendations on how to address psychological issues and make effective referrals in routine practice.

Previous studies had highlighted that the burden of chronic digestive diseases is amplified by psychosocial factors, including poor coping, depression, and poor social support. Mental health professionals specializing in psychogastroenterology integrate the use of brain-gut psychotherapies into GI practice settings, which may help reduce health care utilization and symptom burden.

The article contains best practice advice based on a review of the literature, including existing systematic reviews and expert opinions. These best practices include the following:

  • Gastroenterologists routinely should assess health-related quality of life, symptom-specific anxieties, early-life adversity, and functional impairment related to a patient’s digestive complaints.
  • Gastroenterologists should master patient-friendly language to help explain the brain-gut pathway and how this pathway can become dysregulated by any number of factors, the psychosocial risks perpetuating and maintaining factors of GI diseases, and why the gastroenterologist is referring a patient to a mental health provider.
  • Gastroenterologists should know the structure and core features of the most effective brain-gut psychotherapies.
  • Gastroenterologists should establish a direct referral and ongoing communication pathway with one or two qualified mental health providers and assure patients that he/she will remain a part of the care team.
  • Gastroenterologists should familiarize themselves with one or two neuromodulators that can be used to augment behavioral therapies when necessary.

Patient education about the referral to a mental health provider is difficult and requires attention to detail and fostering a good physician-patient relationship. It is important to help patients understand why they are being referred to a psychologist for a gastrointestinal complaint and that their physical symptoms are not being discounted. Failure to properly explain the reason for referral may lead to poor follow-through and even lead the patient to seek care with another provider.

In order to foster widespread integration of these services, research and clinical gaps need to be addressed. Research gaps include the lack of prospective trials that compare the relative effectiveness of brain-gut psychotherapies with each other and/or with that of psychotropic medications. Other promising brain-gut therapies, such as mindfulness meditation or acceptance-based approaches, lack sufficient research to be included in clinical practice. Limited evidence supports the effect of psychotherapies have in accelerating or enhancing the efficacy of pharmacologic therapies and on improving disease course or inflammation in conditions such as Crohn’s and ulcerative colitis.

 

 


Clinical gaps include the need for better coverage for these therapies by insurance – many providers are out of network or do not accept insurance, although Medicare and commercial insurance plans often cover the cost of services in network. Health psychologists can be reimbursed for health and behavior codes for treating these conditions (CPTs 96150/96152), but there are restrictions on which other types of professionals can use them. Ongoing research is focusing on the cost-effectiveness of these therapies, although some highly effective therapies may be short term and have a one-time total cost of $1,000-$2,000 paid out of pocket. There is a growing need to expand remote, online, or digitally based brain-gut therapies with more trained health care providers that could offset overhead and other therapy costs.
Publications
Topics
Sections

Psychogastroenterology is the science of applying psychological principles and techniques to alleviate the burden of chronic digestive diseases. This burden includes digestive symptoms and disease severity, as well as patients’ ability to cope with them. Chronic digestive diseases, such as irritable bowel syndrome, gastroesophageal reflux disease, and inflammatory bowel diseases, cannot be disentangled from their psychosocial context. In this regard, the role of gastroenterologists in promoting best practices for the assessment and referral of patients across the spectrum of disease to brain-gut psychotherapies is crucial.

A review by Laurie Keefer, PhD, AGAF, and her coauthors, published in the April issue of Gastroenterology, provided a clinical update on the structure and efficacy of two major classes of psychogastroenterology – cognitive-behavioral therapy (CBT) and gut-directed hypnotherapy (HYP). The review discussed the effects of these therapies on GI symptoms and the patients’ ability to improve coping, resilience, and self-regulation. The review also provided a framework to understand the scientific rationale and best practices associated with incorporating brain-gut psychotherapies into routine GI care. Furthermore, it presented recommendations on how to address psychological issues and make effective referrals in routine practice.

Previous studies had highlighted that the burden of chronic digestive diseases is amplified by psychosocial factors, including poor coping, depression, and poor social support. Mental health professionals specializing in psychogastroenterology integrate the use of brain-gut psychotherapies into GI practice settings, which may help reduce health care utilization and symptom burden.

The article contains best practice advice based on a review of the literature, including existing systematic reviews and expert opinions. These best practices include the following:

  • Gastroenterologists routinely should assess health-related quality of life, symptom-specific anxieties, early-life adversity, and functional impairment related to a patient’s digestive complaints.
  • Gastroenterologists should master patient-friendly language to help explain the brain-gut pathway and how this pathway can become dysregulated by any number of factors, the psychosocial risks perpetuating and maintaining factors of GI diseases, and why the gastroenterologist is referring a patient to a mental health provider.
  • Gastroenterologists should know the structure and core features of the most effective brain-gut psychotherapies.
  • Gastroenterologists should establish a direct referral and ongoing communication pathway with one or two qualified mental health providers and assure patients that he/she will remain a part of the care team.
  • Gastroenterologists should familiarize themselves with one or two neuromodulators that can be used to augment behavioral therapies when necessary.

Patient education about the referral to a mental health provider is difficult and requires attention to detail and fostering a good physician-patient relationship. It is important to help patients understand why they are being referred to a psychologist for a gastrointestinal complaint and that their physical symptoms are not being discounted. Failure to properly explain the reason for referral may lead to poor follow-through and even lead the patient to seek care with another provider.

In order to foster widespread integration of these services, research and clinical gaps need to be addressed. Research gaps include the lack of prospective trials that compare the relative effectiveness of brain-gut psychotherapies with each other and/or with that of psychotropic medications. Other promising brain-gut therapies, such as mindfulness meditation or acceptance-based approaches, lack sufficient research to be included in clinical practice. Limited evidence supports the effect of psychotherapies have in accelerating or enhancing the efficacy of pharmacologic therapies and on improving disease course or inflammation in conditions such as Crohn’s and ulcerative colitis.

 

 


Clinical gaps include the need for better coverage for these therapies by insurance – many providers are out of network or do not accept insurance, although Medicare and commercial insurance plans often cover the cost of services in network. Health psychologists can be reimbursed for health and behavior codes for treating these conditions (CPTs 96150/96152), but there are restrictions on which other types of professionals can use them. Ongoing research is focusing on the cost-effectiveness of these therapies, although some highly effective therapies may be short term and have a one-time total cost of $1,000-$2,000 paid out of pocket. There is a growing need to expand remote, online, or digitally based brain-gut therapies with more trained health care providers that could offset overhead and other therapy costs.

Psychogastroenterology is the science of applying psychological principles and techniques to alleviate the burden of chronic digestive diseases. This burden includes digestive symptoms and disease severity, as well as patients’ ability to cope with them. Chronic digestive diseases, such as irritable bowel syndrome, gastroesophageal reflux disease, and inflammatory bowel diseases, cannot be disentangled from their psychosocial context. In this regard, the role of gastroenterologists in promoting best practices for the assessment and referral of patients across the spectrum of disease to brain-gut psychotherapies is crucial.

A review by Laurie Keefer, PhD, AGAF, and her coauthors, published in the April issue of Gastroenterology, provided a clinical update on the structure and efficacy of two major classes of psychogastroenterology – cognitive-behavioral therapy (CBT) and gut-directed hypnotherapy (HYP). The review discussed the effects of these therapies on GI symptoms and the patients’ ability to improve coping, resilience, and self-regulation. The review also provided a framework to understand the scientific rationale and best practices associated with incorporating brain-gut psychotherapies into routine GI care. Furthermore, it presented recommendations on how to address psychological issues and make effective referrals in routine practice.

Previous studies had highlighted that the burden of chronic digestive diseases is amplified by psychosocial factors, including poor coping, depression, and poor social support. Mental health professionals specializing in psychogastroenterology integrate the use of brain-gut psychotherapies into GI practice settings, which may help reduce health care utilization and symptom burden.

The article contains best practice advice based on a review of the literature, including existing systematic reviews and expert opinions. These best practices include the following:

  • Gastroenterologists routinely should assess health-related quality of life, symptom-specific anxieties, early-life adversity, and functional impairment related to a patient’s digestive complaints.
  • Gastroenterologists should master patient-friendly language to help explain the brain-gut pathway and how this pathway can become dysregulated by any number of factors, the psychosocial risks perpetuating and maintaining factors of GI diseases, and why the gastroenterologist is referring a patient to a mental health provider.
  • Gastroenterologists should know the structure and core features of the most effective brain-gut psychotherapies.
  • Gastroenterologists should establish a direct referral and ongoing communication pathway with one or two qualified mental health providers and assure patients that he/she will remain a part of the care team.
  • Gastroenterologists should familiarize themselves with one or two neuromodulators that can be used to augment behavioral therapies when necessary.

Patient education about the referral to a mental health provider is difficult and requires attention to detail and fostering a good physician-patient relationship. It is important to help patients understand why they are being referred to a psychologist for a gastrointestinal complaint and that their physical symptoms are not being discounted. Failure to properly explain the reason for referral may lead to poor follow-through and even lead the patient to seek care with another provider.

In order to foster widespread integration of these services, research and clinical gaps need to be addressed. Research gaps include the lack of prospective trials that compare the relative effectiveness of brain-gut psychotherapies with each other and/or with that of psychotropic medications. Other promising brain-gut therapies, such as mindfulness meditation or acceptance-based approaches, lack sufficient research to be included in clinical practice. Limited evidence supports the effect of psychotherapies have in accelerating or enhancing the efficacy of pharmacologic therapies and on improving disease course or inflammation in conditions such as Crohn’s and ulcerative colitis.

 

 


Clinical gaps include the need for better coverage for these therapies by insurance – many providers are out of network or do not accept insurance, although Medicare and commercial insurance plans often cover the cost of services in network. Health psychologists can be reimbursed for health and behavior codes for treating these conditions (CPTs 96150/96152), but there are restrictions on which other types of professionals can use them. Ongoing research is focusing on the cost-effectiveness of these therapies, although some highly effective therapies may be short term and have a one-time total cost of $1,000-$2,000 paid out of pocket. There is a growing need to expand remote, online, or digitally based brain-gut therapies with more trained health care providers that could offset overhead and other therapy costs.
Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Lower socioeconomic status linked with poor NSCLC prognosis in those with pretreatment weight loss

Article Type
Changed
Fri, 01/04/2019 - 14:14

 

Socioeconomic status, as measured by insurance status, was associated with survival among patients with non–small cell lung cancer (NSCLC) and pretreatment cancer-associated weight loss, in a retrospective review of medical records.

SOURCE: Lau S et al. J Oncol Pract. 2018 Mar 20. doi: 10.1200/JOP.2017.025239

Publications
Topics
Sections

 

Socioeconomic status, as measured by insurance status, was associated with survival among patients with non–small cell lung cancer (NSCLC) and pretreatment cancer-associated weight loss, in a retrospective review of medical records.

SOURCE: Lau S et al. J Oncol Pract. 2018 Mar 20. doi: 10.1200/JOP.2017.025239

 

Socioeconomic status, as measured by insurance status, was associated with survival among patients with non–small cell lung cancer (NSCLC) and pretreatment cancer-associated weight loss, in a retrospective review of medical records.

SOURCE: Lau S et al. J Oncol Pract. 2018 Mar 20. doi: 10.1200/JOP.2017.025239

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM JOURNAL OF ONCOLOGY PRACTICE

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Early recognition and management of cancer-associated weight loss in patients with NSCLC and low socioeconomic status may improve outcomes.

Major finding: Lack of insurance was significantly prognostic among patients with NSCLC and pretreatment weight loss (hazard ratio, 1.63 95% CI, 1.14-2.35).

Study details: 1,366 adult patients with NSCLC consecutively treated at a tertiary care health system between Jan. 1, 2006, and Dec. 31, 2013.

Disclosures: The study was supported in part by National Center for Advancing Translational Sciences grants TL1TR001104 and UL1TR001105. Dr. Lau reported employment with LabCorp by an immediate family member. Coauthors reported financial ties to Advenchen Laboratories, Macrogen, Peregrine Pharmaceuticals, and DFINE.

Source: Lau S et al. J Oncol Pract. 2018 Mar 20. doi: 10.1200/JOP.2017.025239.

Disqus Comments
Default

Bioengineered liver models screen drugs and study liver injury

Article Type
Changed
Sat, 12/08/2018 - 14:51

Bioengineered liver models have enabled recapitulation of liver architecture with precise control over cellular microenvironments, resulting in stabilized liver functions for several weeks in vitro. Studies have focused on using these models to investigate cell responses to drugs and other stimuli (for example, viruses and cell differentiation cues) to predict clinical outcomes. Gregory H. Underhill, PhD, from the department of bioengineering at the University of Illinois at Urbana-Champaign and Salman R. Khetani, PhD, from the department of bioengineering at the University of Illinois in Chicago presented a comprehensive review of the these advances in bioengineered liver models in Cellular and Molecular Gastroenterology and Hepatology (doi: 10.1016/j.jcmgh.2017.11.012).

Drug-induced liver injury (DILI) is a leading cause of drug attrition in the United States, with some marketed drugs causing cell necrosis, hepatitis, cholestasis, fibrosis, or a mixture of injury types. Although the Food and Drug Administration requires preclinical drug testing in animal models, differences in species-specific drug metabolism pathways and human genetics may result in inadequate identification of potential for human DILI. Some bioengineered liver models for in vitro studies are based on tissue engineering using high-throughput microarrays, protein micropatterning, microfluidics, specialized plates, biomaterial scaffolds, and bioprinting.

High-throughput cell microarrays enable systematic analysis of a large number of drugs or compounds at a relatively low cost. Several culture platforms have been developed using multiple sources of liver cells, including cancerous and immortalized cell lines. These platforms show enhanced capabilities to evaluate combinatorial effects of multiple signals with independent control of biochemical and biomechanical cues. For instance, a microchip platform for transducing 3-D liver cell cultures with genes for drug metabolism enzymes featuring 532 reaction vessels (micropillars and corresponding microwells) was able to provide information about certain enzyme combinations that led to drug toxicity in cells. The high-throughput cell microarrays are, however, primarily dependent on imaging-based readouts and have a limited ability to investigate cell responses to gradients of microenvironmental signals.

Liver development, physiology, and pathophysiology are dependent on homotypic and heterotypic interactions between parenchymal and nonparenchymal cells (NPCs). Cocultures with both liver- and nonliver-derived NPC types, in vitro, can induce liver functions transiently and have proven useful for investigating host responses to sepsis, mutagenesis, xenobiotic metabolism and toxicity, response to oxidative stress, lipid metabolism, and induction of the acute-phase response. Micropatterned cocultures (MPCCs) are designed to allow the use of different NPC types without significantly altering hepatocyte homotypic interactions. Cell-cell interactions can be precisely controlled to allow for stable functions for up to 4-6 weeks, whereas more randomly distributed cocultures have limited stability. Unlike randomly distributed cocultures, MPCCs can be infected with HBV, HCV, and malaria. Potential limitations of MPCCs include the requirement for specialized equipment and devices for patterning collagen for hepatocyte attachment.

 

 


Randomly distributed spheroids or organoids enable 3-D establishment of homotypic cell-cell interactions surrounded by an extracellular matrix. The spheroids can be further cocultured with NPCs that facilitate heterotypic cell-cell interactions and allow the evaluation of outcomes resulting from drugs and other stimuli. Hepatic spheroids maintain major liver functions for several weeks and have proven to be compatible with multiple applications within the drug development pipeline.

These spheroids showed greater sensitivity in identifying known hepatotoxic drugs than did short-term primary human hepatocyte (PHH) monolayers. PHHs secreted liver proteins, such as albumin, transferrin, and fibrinogen, and showed cytochrome-P450 activities for 77-90 days when cultured on a nylon scaffold containing a mixture of liver NPCs and PHHs.

Nanopillar plates can be used to create induced pluripotent stem cell–derived human hepatocyte-like cell (iHep) spheroids; although these spheroids showed some potential for initial drug toxicity screening, they had lower overall sensitivity than conventional PHH monolayers, which suggests that further maturation of iHeps is likely required.

Potential limitations of randomly distributed spheroids include necrosis of cells in the center of larger spheroids and the requirement for expensive confocal microscopy for high-content imaging of entire spheroid cultures. To overcome the limitation of disorganized cell type interactions over time within the randomly distributed spheroids/organoids, bioprinted human liver organoids are designed to allow precise control of cell placement.

 

 

SOURCE: Underhill GH and Khetani SR. Cell Molec Gastro Hepatol. 2017. doi: org/10.1016/j.jcmgh.2017.11.012.

Body

 

Thirty to 50 new drugs are approved in the United States annually, which costs approximately $2.5 billion/drug in drug development costs. Nine out of 10 drugs never make it to market, and of those that do, adverse events affect their longevity. Hepatotoxicity is the most frequent adverse drug reaction, and drug-induced liver injury, which can lead to acute liver failure, occurs in a subset of affected patients. Understanding a drug’s risk of hepatotoxicity before patients start using it can not only save lives but also conceivably reduce the costs incurred by pharmaceutical companies, which are passed on to consumers.

Dr. Rotonya Carr Assistant professor of medicine, division of gastroenterology, University of Pennsylvania
Dr. Rotonya Carr
In Cellular and Molecular Gastroenterology and Hepatology, Underhill and Khetani summarize available and emerging cell-based, high-throughput systems that can be used to predict hepatotoxicity. These modalities include cellular microarrays of single cells; cocultures of liver parenchymal and nonparenchymal cells; organoids (3-D organ-like structures); and liver-on-a-chip devices (complex perfusion bioreactors that allow for modulation of the cellular micro-environment). These in vitro systems have not only enabled investigators to screen multiple drugs at the same time but also have informed the clinical translation of these technologies. For example, the extracorporeal liver assist device – essentially, a liver bypass – and similar bioartificial liver devices can in principal temporarily perform some of the major liver functions while a patient’s native liver heals from drug-induced liver injury or other hepatic injury.

However, just as we have seen with the limitations of the in vitro systems, bioartificial livers are unlikely to be successful unless they integrate the liver’s complex functions of protein synthesis, immune surveillance, energy homeostasis, and nutrient sensing. The future is bright, though, as biomedical scientists and bioengineers continue to push the envelope by advancing both in vitro and bioartificial technologies.

Rotonya Carr, MD, is an assistant professor of medicine in the division of gastroenterology at the University of Pennsylvania, Philadelphia. She receives research support from Intercept Pharmaceuticals.

Publications
Topics
Sections
Body

 

Thirty to 50 new drugs are approved in the United States annually, which costs approximately $2.5 billion/drug in drug development costs. Nine out of 10 drugs never make it to market, and of those that do, adverse events affect their longevity. Hepatotoxicity is the most frequent adverse drug reaction, and drug-induced liver injury, which can lead to acute liver failure, occurs in a subset of affected patients. Understanding a drug’s risk of hepatotoxicity before patients start using it can not only save lives but also conceivably reduce the costs incurred by pharmaceutical companies, which are passed on to consumers.

Dr. Rotonya Carr Assistant professor of medicine, division of gastroenterology, University of Pennsylvania
Dr. Rotonya Carr
In Cellular and Molecular Gastroenterology and Hepatology, Underhill and Khetani summarize available and emerging cell-based, high-throughput systems that can be used to predict hepatotoxicity. These modalities include cellular microarrays of single cells; cocultures of liver parenchymal and nonparenchymal cells; organoids (3-D organ-like structures); and liver-on-a-chip devices (complex perfusion bioreactors that allow for modulation of the cellular micro-environment). These in vitro systems have not only enabled investigators to screen multiple drugs at the same time but also have informed the clinical translation of these technologies. For example, the extracorporeal liver assist device – essentially, a liver bypass – and similar bioartificial liver devices can in principal temporarily perform some of the major liver functions while a patient’s native liver heals from drug-induced liver injury or other hepatic injury.

However, just as we have seen with the limitations of the in vitro systems, bioartificial livers are unlikely to be successful unless they integrate the liver’s complex functions of protein synthesis, immune surveillance, energy homeostasis, and nutrient sensing. The future is bright, though, as biomedical scientists and bioengineers continue to push the envelope by advancing both in vitro and bioartificial technologies.

Rotonya Carr, MD, is an assistant professor of medicine in the division of gastroenterology at the University of Pennsylvania, Philadelphia. She receives research support from Intercept Pharmaceuticals.

Body

 

Thirty to 50 new drugs are approved in the United States annually, which costs approximately $2.5 billion/drug in drug development costs. Nine out of 10 drugs never make it to market, and of those that do, adverse events affect their longevity. Hepatotoxicity is the most frequent adverse drug reaction, and drug-induced liver injury, which can lead to acute liver failure, occurs in a subset of affected patients. Understanding a drug’s risk of hepatotoxicity before patients start using it can not only save lives but also conceivably reduce the costs incurred by pharmaceutical companies, which are passed on to consumers.

Dr. Rotonya Carr Assistant professor of medicine, division of gastroenterology, University of Pennsylvania
Dr. Rotonya Carr
In Cellular and Molecular Gastroenterology and Hepatology, Underhill and Khetani summarize available and emerging cell-based, high-throughput systems that can be used to predict hepatotoxicity. These modalities include cellular microarrays of single cells; cocultures of liver parenchymal and nonparenchymal cells; organoids (3-D organ-like structures); and liver-on-a-chip devices (complex perfusion bioreactors that allow for modulation of the cellular micro-environment). These in vitro systems have not only enabled investigators to screen multiple drugs at the same time but also have informed the clinical translation of these technologies. For example, the extracorporeal liver assist device – essentially, a liver bypass – and similar bioartificial liver devices can in principal temporarily perform some of the major liver functions while a patient’s native liver heals from drug-induced liver injury or other hepatic injury.

However, just as we have seen with the limitations of the in vitro systems, bioartificial livers are unlikely to be successful unless they integrate the liver’s complex functions of protein synthesis, immune surveillance, energy homeostasis, and nutrient sensing. The future is bright, though, as biomedical scientists and bioengineers continue to push the envelope by advancing both in vitro and bioartificial technologies.

Rotonya Carr, MD, is an assistant professor of medicine in the division of gastroenterology at the University of Pennsylvania, Philadelphia. She receives research support from Intercept Pharmaceuticals.

Bioengineered liver models have enabled recapitulation of liver architecture with precise control over cellular microenvironments, resulting in stabilized liver functions for several weeks in vitro. Studies have focused on using these models to investigate cell responses to drugs and other stimuli (for example, viruses and cell differentiation cues) to predict clinical outcomes. Gregory H. Underhill, PhD, from the department of bioengineering at the University of Illinois at Urbana-Champaign and Salman R. Khetani, PhD, from the department of bioengineering at the University of Illinois in Chicago presented a comprehensive review of the these advances in bioengineered liver models in Cellular and Molecular Gastroenterology and Hepatology (doi: 10.1016/j.jcmgh.2017.11.012).

Drug-induced liver injury (DILI) is a leading cause of drug attrition in the United States, with some marketed drugs causing cell necrosis, hepatitis, cholestasis, fibrosis, or a mixture of injury types. Although the Food and Drug Administration requires preclinical drug testing in animal models, differences in species-specific drug metabolism pathways and human genetics may result in inadequate identification of potential for human DILI. Some bioengineered liver models for in vitro studies are based on tissue engineering using high-throughput microarrays, protein micropatterning, microfluidics, specialized plates, biomaterial scaffolds, and bioprinting.

High-throughput cell microarrays enable systematic analysis of a large number of drugs or compounds at a relatively low cost. Several culture platforms have been developed using multiple sources of liver cells, including cancerous and immortalized cell lines. These platforms show enhanced capabilities to evaluate combinatorial effects of multiple signals with independent control of biochemical and biomechanical cues. For instance, a microchip platform for transducing 3-D liver cell cultures with genes for drug metabolism enzymes featuring 532 reaction vessels (micropillars and corresponding microwells) was able to provide information about certain enzyme combinations that led to drug toxicity in cells. The high-throughput cell microarrays are, however, primarily dependent on imaging-based readouts and have a limited ability to investigate cell responses to gradients of microenvironmental signals.

Liver development, physiology, and pathophysiology are dependent on homotypic and heterotypic interactions between parenchymal and nonparenchymal cells (NPCs). Cocultures with both liver- and nonliver-derived NPC types, in vitro, can induce liver functions transiently and have proven useful for investigating host responses to sepsis, mutagenesis, xenobiotic metabolism and toxicity, response to oxidative stress, lipid metabolism, and induction of the acute-phase response. Micropatterned cocultures (MPCCs) are designed to allow the use of different NPC types without significantly altering hepatocyte homotypic interactions. Cell-cell interactions can be precisely controlled to allow for stable functions for up to 4-6 weeks, whereas more randomly distributed cocultures have limited stability. Unlike randomly distributed cocultures, MPCCs can be infected with HBV, HCV, and malaria. Potential limitations of MPCCs include the requirement for specialized equipment and devices for patterning collagen for hepatocyte attachment.

 

 


Randomly distributed spheroids or organoids enable 3-D establishment of homotypic cell-cell interactions surrounded by an extracellular matrix. The spheroids can be further cocultured with NPCs that facilitate heterotypic cell-cell interactions and allow the evaluation of outcomes resulting from drugs and other stimuli. Hepatic spheroids maintain major liver functions for several weeks and have proven to be compatible with multiple applications within the drug development pipeline.

These spheroids showed greater sensitivity in identifying known hepatotoxic drugs than did short-term primary human hepatocyte (PHH) monolayers. PHHs secreted liver proteins, such as albumin, transferrin, and fibrinogen, and showed cytochrome-P450 activities for 77-90 days when cultured on a nylon scaffold containing a mixture of liver NPCs and PHHs.

Nanopillar plates can be used to create induced pluripotent stem cell–derived human hepatocyte-like cell (iHep) spheroids; although these spheroids showed some potential for initial drug toxicity screening, they had lower overall sensitivity than conventional PHH monolayers, which suggests that further maturation of iHeps is likely required.

Potential limitations of randomly distributed spheroids include necrosis of cells in the center of larger spheroids and the requirement for expensive confocal microscopy for high-content imaging of entire spheroid cultures. To overcome the limitation of disorganized cell type interactions over time within the randomly distributed spheroids/organoids, bioprinted human liver organoids are designed to allow precise control of cell placement.

 

 

SOURCE: Underhill GH and Khetani SR. Cell Molec Gastro Hepatol. 2017. doi: org/10.1016/j.jcmgh.2017.11.012.

Bioengineered liver models have enabled recapitulation of liver architecture with precise control over cellular microenvironments, resulting in stabilized liver functions for several weeks in vitro. Studies have focused on using these models to investigate cell responses to drugs and other stimuli (for example, viruses and cell differentiation cues) to predict clinical outcomes. Gregory H. Underhill, PhD, from the department of bioengineering at the University of Illinois at Urbana-Champaign and Salman R. Khetani, PhD, from the department of bioengineering at the University of Illinois in Chicago presented a comprehensive review of the these advances in bioengineered liver models in Cellular and Molecular Gastroenterology and Hepatology (doi: 10.1016/j.jcmgh.2017.11.012).

Drug-induced liver injury (DILI) is a leading cause of drug attrition in the United States, with some marketed drugs causing cell necrosis, hepatitis, cholestasis, fibrosis, or a mixture of injury types. Although the Food and Drug Administration requires preclinical drug testing in animal models, differences in species-specific drug metabolism pathways and human genetics may result in inadequate identification of potential for human DILI. Some bioengineered liver models for in vitro studies are based on tissue engineering using high-throughput microarrays, protein micropatterning, microfluidics, specialized plates, biomaterial scaffolds, and bioprinting.

High-throughput cell microarrays enable systematic analysis of a large number of drugs or compounds at a relatively low cost. Several culture platforms have been developed using multiple sources of liver cells, including cancerous and immortalized cell lines. These platforms show enhanced capabilities to evaluate combinatorial effects of multiple signals with independent control of biochemical and biomechanical cues. For instance, a microchip platform for transducing 3-D liver cell cultures with genes for drug metabolism enzymes featuring 532 reaction vessels (micropillars and corresponding microwells) was able to provide information about certain enzyme combinations that led to drug toxicity in cells. The high-throughput cell microarrays are, however, primarily dependent on imaging-based readouts and have a limited ability to investigate cell responses to gradients of microenvironmental signals.

Liver development, physiology, and pathophysiology are dependent on homotypic and heterotypic interactions between parenchymal and nonparenchymal cells (NPCs). Cocultures with both liver- and nonliver-derived NPC types, in vitro, can induce liver functions transiently and have proven useful for investigating host responses to sepsis, mutagenesis, xenobiotic metabolism and toxicity, response to oxidative stress, lipid metabolism, and induction of the acute-phase response. Micropatterned cocultures (MPCCs) are designed to allow the use of different NPC types without significantly altering hepatocyte homotypic interactions. Cell-cell interactions can be precisely controlled to allow for stable functions for up to 4-6 weeks, whereas more randomly distributed cocultures have limited stability. Unlike randomly distributed cocultures, MPCCs can be infected with HBV, HCV, and malaria. Potential limitations of MPCCs include the requirement for specialized equipment and devices for patterning collagen for hepatocyte attachment.

 

 


Randomly distributed spheroids or organoids enable 3-D establishment of homotypic cell-cell interactions surrounded by an extracellular matrix. The spheroids can be further cocultured with NPCs that facilitate heterotypic cell-cell interactions and allow the evaluation of outcomes resulting from drugs and other stimuli. Hepatic spheroids maintain major liver functions for several weeks and have proven to be compatible with multiple applications within the drug development pipeline.

These spheroids showed greater sensitivity in identifying known hepatotoxic drugs than did short-term primary human hepatocyte (PHH) monolayers. PHHs secreted liver proteins, such as albumin, transferrin, and fibrinogen, and showed cytochrome-P450 activities for 77-90 days when cultured on a nylon scaffold containing a mixture of liver NPCs and PHHs.

Nanopillar plates can be used to create induced pluripotent stem cell–derived human hepatocyte-like cell (iHep) spheroids; although these spheroids showed some potential for initial drug toxicity screening, they had lower overall sensitivity than conventional PHH monolayers, which suggests that further maturation of iHeps is likely required.

Potential limitations of randomly distributed spheroids include necrosis of cells in the center of larger spheroids and the requirement for expensive confocal microscopy for high-content imaging of entire spheroid cultures. To overcome the limitation of disorganized cell type interactions over time within the randomly distributed spheroids/organoids, bioprinted human liver organoids are designed to allow precise control of cell placement.

 

 

SOURCE: Underhill GH and Khetani SR. Cell Molec Gastro Hepatol. 2017. doi: org/10.1016/j.jcmgh.2017.11.012.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Gaps exist in receipt of clinically indicated genetic counseling after breast cancer diagnosis

Article Type
Changed
Thu, 12/15/2022 - 17:48

 

A minority of patients (43.5%) with an indication for breast cancer genetic risk evaluation actually received formal genetic counseling in clinical practice, according to an analysis of NCI Surveillance, Epidemiology, and End Results (SEER) data published in Journal of Clinical Oncology.

More expertise is required in genetic counseling, either formal counseling given by an expert, or by a cancer physician (physician-directed), wrote Steven J. Katz and his colleagues at the University of Michigan, Ann Arbor. With BRCA1/2-only testing, being replaced by multi-gene panel testing, further consideration and/or discussion of results and formulation of a management plan is required, they said.

Dr. Steven J. Katz of the University of Michigan, Ann Arbor
Dr. Steven J. Katz
Of 5,080 women with favorable breast cancer prognosis identified in the SEER registries between 2013 and 2015 in Georgia and Los Angeles County, 1,171 were identified as having clinical indications for formal genetic risk evaluation according to NCCN guidelines.

Of those, 47.4% did not get tested, 40.7% tested negative, 7.4% had a variant of uncertain significance only, and 4.5% had a pathogenic mutation. Three quarters (74.6%) received some form of genetic counseling (43.5%, formal counseling and 31.1%, physician-directed discussion). Almost all tested patients (96.1%) reported some form of genetic discussion. One half (50.6%) of those not tested received any discussion about genetics, reported the authors.

 

 

SOURCE: Katz SJ et al. J Clin Oncol. 2018 Mar 12. doi: 10.1200/JCO.2017.76.2369.

Publications
Topics
Sections

 

A minority of patients (43.5%) with an indication for breast cancer genetic risk evaluation actually received formal genetic counseling in clinical practice, according to an analysis of NCI Surveillance, Epidemiology, and End Results (SEER) data published in Journal of Clinical Oncology.

More expertise is required in genetic counseling, either formal counseling given by an expert, or by a cancer physician (physician-directed), wrote Steven J. Katz and his colleagues at the University of Michigan, Ann Arbor. With BRCA1/2-only testing, being replaced by multi-gene panel testing, further consideration and/or discussion of results and formulation of a management plan is required, they said.

Dr. Steven J. Katz of the University of Michigan, Ann Arbor
Dr. Steven J. Katz
Of 5,080 women with favorable breast cancer prognosis identified in the SEER registries between 2013 and 2015 in Georgia and Los Angeles County, 1,171 were identified as having clinical indications for formal genetic risk evaluation according to NCCN guidelines.

Of those, 47.4% did not get tested, 40.7% tested negative, 7.4% had a variant of uncertain significance only, and 4.5% had a pathogenic mutation. Three quarters (74.6%) received some form of genetic counseling (43.5%, formal counseling and 31.1%, physician-directed discussion). Almost all tested patients (96.1%) reported some form of genetic discussion. One half (50.6%) of those not tested received any discussion about genetics, reported the authors.

 

 

SOURCE: Katz SJ et al. J Clin Oncol. 2018 Mar 12. doi: 10.1200/JCO.2017.76.2369.

 

A minority of patients (43.5%) with an indication for breast cancer genetic risk evaluation actually received formal genetic counseling in clinical practice, according to an analysis of NCI Surveillance, Epidemiology, and End Results (SEER) data published in Journal of Clinical Oncology.

More expertise is required in genetic counseling, either formal counseling given by an expert, or by a cancer physician (physician-directed), wrote Steven J. Katz and his colleagues at the University of Michigan, Ann Arbor. With BRCA1/2-only testing, being replaced by multi-gene panel testing, further consideration and/or discussion of results and formulation of a management plan is required, they said.

Dr. Steven J. Katz of the University of Michigan, Ann Arbor
Dr. Steven J. Katz
Of 5,080 women with favorable breast cancer prognosis identified in the SEER registries between 2013 and 2015 in Georgia and Los Angeles County, 1,171 were identified as having clinical indications for formal genetic risk evaluation according to NCCN guidelines.

Of those, 47.4% did not get tested, 40.7% tested negative, 7.4% had a variant of uncertain significance only, and 4.5% had a pathogenic mutation. Three quarters (74.6%) received some form of genetic counseling (43.5%, formal counseling and 31.1%, physician-directed discussion). Almost all tested patients (96.1%) reported some form of genetic discussion. One half (50.6%) of those not tested received any discussion about genetics, reported the authors.

 

 

SOURCE: Katz SJ et al. J Clin Oncol. 2018 Mar 12. doi: 10.1200/JCO.2017.76.2369.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM JOURNAL OF CLINICAL ONCOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: There exists a large gap between mandates for timely pretest formal genetic counseling of higher-risk, breast cancer patients and its implementation in clinical practice.

Major finding: Almost half (47.4%) of patients diagnosed with early breast cancer with an indication for genetic risk evaluation did not get genetic tests. Of those who got genetic testing, 43.5% received formal counseling and 31.1% received physician-directed discussion.

Study details: Data on 5,080 women aged 20-79 years diagnosed with early stage breast during 2013-2015, reported to National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) registries of Georgia and Los Angeles County.

Disclosures: Potential conflict of interests were reported by Lauren P. Wallner, PhD (GlaxoSmithKline); Monica Morrow, MD (Genomic Health); Reshma Jagsi, MD (Amgen and AbbVie); and Allison W. Kurian, MD (Myriad Genetics, Invitae, Ambry Genetics, Genomic Health, GeneDx/BioReference, Genentech (a member of the Roche Group).

Source: Katz SJ et al. J Clin Oncol. 2018 Mar 12. doi: 10.1200/JCO.2017.76.2369.

Disqus Comments
Default

Racial disparities by region persist despite multiple liver transplant allocation schemes

Article Type
Changed
Wed, 01/02/2019 - 10:06

 

Racial and regional disparities in liver transplant allocation persist despite multiple allocation schemes as identified in a large dataset spanning at least 30 years, according to a study.

An operation in the OR
VILevi/Thinkstock

 

 

SOURCE: Monlezun DJ et al. Surgery. 2018 doi: 10.1016/j.surg.2017.10.009.

Publications
Topics
Sections

 

Racial and regional disparities in liver transplant allocation persist despite multiple allocation schemes as identified in a large dataset spanning at least 30 years, according to a study.

An operation in the OR
VILevi/Thinkstock

 

 

SOURCE: Monlezun DJ et al. Surgery. 2018 doi: 10.1016/j.surg.2017.10.009.

 

Racial and regional disparities in liver transplant allocation persist despite multiple allocation schemes as identified in a large dataset spanning at least 30 years, according to a study.

An operation in the OR
VILevi/Thinkstock

 

 

SOURCE: Monlezun DJ et al. Surgery. 2018 doi: 10.1016/j.surg.2017.10.009.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM SURGERY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: The existence of racial disparity in liver allograft distribution is undisputed. Disagreements persist over optimal allocation schemes, with centers using different schemes.

Major finding: A rigorous causal inference statistic on a large national dataset spanning at least 30 years showed that racial disparities by region persist despite multiple allocation schemes.

Study details: Patients from the United Network for Organ Sharing Standard Transplant Analysis and Research database (1985-2016) were used to assess causal inference of racial and regional disparities.

Disclosures: None reported.

Source: Monlezun DJ et al. Surgery. 2018. doi: 10.1016/j.surg.2017.10.009.

Disqus Comments
Default