throbber
Editorials
`
`Teduglutide and Short Bowel Syndrome: Every Night Without Parenteral
`Fluids Is a Good Night
`
`See “Teduglutide reduces need for parenteral
`support among patients with short bowel syn-
`drome with intestinal failure,” by Jeppesen PB,
`Pertkiewicz M, Messing B, et al, on page 1473.
`
`Short bowel syndrome (SBS) is characterized by either
`
`a congenital or acquired absence of a substantial
`portion of the small intestine. Those patients whose re-
`sultant malabsorption is insufficient to maintain nutri-
`tional or fluid autonomy (eg, fecal energy and/or fluid
`losses are greater than absorption) are deemed to have
`intestinal failure (IF). Although most nutrient absorption
`occurs within the proximal 100 –150 cm of jejunum,1
`intestinal transit time is very rapid, allowing for limited
`nutrient– epithelial contact time. Such individuals are
`among the most complex and challenging to manage
`patients of any gastrointestinal disease. Particularly diffi-
`cult to manage are those patients with a proximal jeju-
`nostomy, who may actually secrete more fluid than they
`ingest.2 Adult patients typically have ⬍200 cm of residual
`small bowel, with or without their colon, although ab-
`sorption is dependent not only on the length of bowel,
`but the overall surface area and function. Underlying
`etiologies may include multiple small bowel resections for
`diseased or obstructed bowel including Crohn’s disease,
`trauma, mesenteric vascular catastrophes, or volvulus. Eti-
`ologies in children may also include congenital disorders
`such as intestinal atresia, malrotation, and gastroschisis.
`In part, because the underlying diseases resulting in SBS
`are a heterogeneous group, there is no ICD-9 code and, as
`such, there are no reliable estimates as to the number of
`individuals with SBS or SBS/IF in the United States.
`Estimates based on European registries and other data-
`bases have suggested the number of such patients in the
`United States may be in the 10,000 –20,000 range.3
`Patients with SBS/IF depend on artificial nutrition
`and/or fluid support to maintain life. This may require
`them to infuse parenteral fluids (parenteral support [PS])
`overnight for 3–7 days per week, and in some cases,
`during the day as well. This therapy has substantial im-
`plications for employment, activities, sleep, and finances.
`Furthermore, medical management may differ depending
`on whether a given patient has residual colon in continu-
`ity with the remaining small intestine or not.
`Numerous complications from SBS or its therapy may
`develop. Most notorious among these is IF-associated
`liver disease.4 IF-associated liver disease is the leading
`indication for intestine and intestine–liver transplanta-
`
`GASTROENTEROLOGY 2012;143:1416 –1434
`
`tion, a procedure that may be life-saving and eliminate
`dependence on parenteral fluids, but may cost upwards of
`$1 million, although often $250,000 –$500,000 for the
`procedure and initial hospitalization in addition to med-
`ications, follow-up clinical visits, and treatment of com-
`plications such as infections or acute and chronic rejec-
`tion and additional surgery. One year of parenteral
`nutrition (PN), discounting treatment for laboratory
`studies, nursing and physician visits, and treatment of
`complications and hospitalizations (generally 1–2 times
`annually), often costs the health care system $100,000 –
`$125,000.5
`Animal models suggest that, after massive intestinal
`resection, the intestine begins to adapt, whereby the in-
`testine lengthens a modest amount, but may increase in
`diameter and surface area significantly. This process is
`more pronounced in the ileum after a jejunal resection
`wherein villi lengthen and crypts deepen.6-8 These findings
`are supported by limited human data,9-12 wherein diar-
`rhea decreases over time after onset of SBS.10 In fact,
`conventional management13 of these patients, perhaps
`not always widely practiced owing to its relative complex-
`ity and unfamiliarity among clinicians,14 combined with
`the patient’s own innate ability for adaptation, can lead to
`elimination of PN in 50% of patients within 6 months of
`their resection.15
`Growth hormone was the first medication approved by
`the US Food and Drug Administration (in 2003) specifi-
`cally for use in patients with SBS receiving specialized
`nutritional support “in conjunction with optimal man-
`agement”13 based on a 2-center controlled trial, although
`there was no placebo group and subjects received addi-
`tional interventions in addition to growth hormone. In-
`consistent results have been reported in previous clinical
`trials. Nevertheless, PN requirements could be reduced by
`approximately 2 L or 1 night weekly.16 The use of this
`growth factor has been limited, largely owing to concerns
`with regard to efficacy and the fact that only short-term
`use was approved. The effects of growth hormone on
`human intestinal absorption are unknown, although it
`enhances reabsorption of sodium in the distal nephron.17
`Notably, patients with acromegaly have a heightened risk
`for development of colonic adenomas in humans, al-
`though malignancy has not been reported.18
`A myriad of other growth factors may be involved in the
`process of postresection intestinal adaptation, including
`hepatocyte growth factor, vascular endothelial growth fac-
`tor, cholecystokinin, transforming growth factor, epider-
`mal growth factor, gastrin, insulin, insulin-like growth
`factor-1, neurotensin, keratinocyte growth factor (KGF),
`
`Page 1
`
`

`
`and glucagon-like peptide-2 (GLP-2).19 GLP-2’s own ac-
`tions may be modulated by some of these peptides, in-
`cluding transforming growth factor-␤, insulin-like growth
`factor-1, and KGF.20-22 Each of these extracellular growth
`factors have the potential for human use. Epidermal
`growth factor is no longer commercially manufactured
`and KGF is used only for the treatment of mucositis
`currently. GLP-2 is among the first of these peptides
`evaluated in humans with SBS/IF.
`GLP-2 is released from L cells in the distal small bowel
`and colon in response to food ingestion, but its release is
`severely blunted in patients with SBS/IF and ileal resec-
`tion,23 although meal-stimulated release is enhanced in
`patients with a preserved colon.24 It promotes intestinal
`epithelial growth via increased cellular proliferation
`through activation of the Wnt signaling pathway, which
`leads to nuclear translocation of ␤-catenin,25 and de-
`creased apoptosis and epithelial growth may also be en-
`hanced through increased mesenteric blood flow.26
`Normal digestion and absorption depends on a gradual
`emptying of nutrients from the stomach into the small
`intestine, wherein mixing with pancreatic enzymes and
`bile occurs. Rapid gastric emptying may result in inade-
`quate mixing, insufficient enzymatic digestion, and im-
`paired nutrient digestion. GLP-2 increases gastrointesti-
`nal transit time, and this may be among the mechanisms
`by which its use leads to decreased diarrhea.27 The use of
`native GLP-2 has resulted in less chronic dehydration, the
`major factor in the development of IF-associated nephrop-
`athy in patients that require long-term PN,28 among patients
`with SBS.29 GLP-2 may also exert a beneficial effect on bone
`health as well,30 which is adversely affected during SBS/IF.31
`However, the actions of the native hormone are limited
`by rapid degradation by dipeptidypeptidase IV, leading
`to a very short half-life. A long-acting analog, more
`resistant to this enzyme, h(Gly2)GLP-2[1-33] (teduglu-
`tide) was developed by the substitution of a glycine
`residue for an alanine in position 2.
`In this issue of GASTROENTEROLOGY, Jeppesen et al32
`report the results of a phase III double-blind, placebo-
`controlled trial of teduglutide for the purposes of enhanc-
`ing nutrient and fluid absorption and weaning parenteral
`fluids in 86 patients with SBS/IF. This study was con-
`ceived because of discrepant results from an earlier place-
`bo-controlled multinational study wherein a higher dose
`of teduglutide (0.1 mg/kg per day) seemed to be slightly
`less effective than the 0.05 mg/kg dose used in the current
`study, although a significant difference for the lower dose
`was not achieved when compared with placebo.33
`Jeppesen et al32 are to be congratulated for completing
`an extremely complex, multinational study in a very com-
`plicated group of patients, in fact the largest prospective
`study ever in this patient population. Patients were re-
`quired to have received PS ⱖ3 times weekly and for a
`
`Editorials, continued
`
`minimum of 12 months immediately preceding the study
`to qualify for inclusion. This could have included PN or
`intravenous fluid and electrolytes alone. A strength of the
`study was the pretreatment PS optimization whereby in-
`dividual patient fluid volumes were stabilized to achieve a
`urine output of 1–2 L/day over a period of 4 –16 weeks
`before randomization. That ensured as much as possible
`that the baseline PS volume was required and stable,
`although unlike in the previous study,33 there was no
`specific attempt at weaning PN before randomization.
`In the current study, patients were randomized to re-
`ceive teduglutide 0.05 mg/kg per day or placebo injected
`daily. The primary endpoint was the percentage of en-
`rolled patients that achieved a consistent reduction in PS
`volume of 20%–100% from baseline at both week 20 and
`week 24. A 20% decrease equates to the reduction of PS
`from 6 to 5 nights per week, a very profound improve-
`ment for an individual patient. Given the disparity be-
`tween daily fluid volumes (ⱖ1.5– 8 L daily), it would have
`been inappropriate to have used the absolute decrease in
`number of days of infusion as the primary endpoint.
`Secondary endpoints in the study included the mean
`percentage and absolute volume decrease in PS and the
`number of patients and the time they required on study
`medication to be completely weaned from PS.
`Attempts to wean PS were made at the discretion of
`individual investigators in increments of 10%–30%, begin-
`ning after 2 weeks of therapy, and then every 4 weeks for
`the remainder of the 24 week study. As we generally do in
`clinical practice, urine output was collected for 48 hours
`(including a night when PS were not administered if ⬍7
`nights per week) before each study visit. This volume had
`to exceed the baseline value before further weaning would
`be considered. Discontinuing an entire day of PS was
`considered based on the weekly volume reduction, but the
`choice was left up to the investigator and patient.
`There were 27 of 43 (63%) responders in the teduglutide
`group compared with 13 of 43 (30%) in the placebo group
`(P ⬍ .02) with differences between the group-specific
`responder rate at each visit. There was a nonsignificant
`trend toward a greater response rate in those patients with
`residual colon in continuity with their remaining small
`bowel. PS volume reduction was 4.4 ⫾ 3.8 L per week in
`the teduglutide group versus 2.3 ⫾ 2.7 L per week in the
`placebo group. At baseline, subjects received an average of
`nearly 2 L/d, so it was not surprising that no subjects were
`completely weaned from PS; however, the number of
`patients who achieved ⱖ1 full day off PS in the teduglu-
`tide group was more than double that in the placebo
`group (54% vs 23%; P ⬍ .047). PS was not completely
`eliminated in any patient. An earlier and more aggres-
`sive PS weaning protocol than that utilized in the
`previous teduglutide study33 may have accounted for a
`
`1417
`
`Page 2
`
`

`
`Editorials, continued
`
`more significant decline in PS volume in the current
`investigations.
`Consistent with an increase in intestinal mass, plasma
`citrulline concentration increased significantly only in the
`teduglutide group. Previous studies with teduglutide have
`shown it reduces the volume of diarrhea,34 although this
`effect was not evaluated in the most recent investiga-
`tion.32 It is to be noted that some patients with SBS/IF
`have sufficient nutrient absorption, but are unable to
`consume sufficient fluids to make up for dramatic losses,
`and as such require parenteral fluid and electrolytes, but
`not PN. Might these patients respond better to teduglu-
`tide? The data were not broken out. In addition, the effect
`of teduglutide on micronutrient absorption and potential
`independence from intravenous micronutrient supple-
`mentation was not evaluated.
`Some patients failed to respond to teduglutide. Why
`did that occur? Because most L cells are located in the
`ileum and colon, patients with resection of those portions
`of bowel would presumably have lower native meal-stim-
`ulated serum GLP-2 concentrations and thereby might
`have a greater response to exogenous administration.
`However, there was a trend toward a greater response in
`patients with colon, although this difference did not at-
`tain significance. Given that nutrient absorption is a fac-
`tor of not only percentage absorption, but oral intake,
`might teduglutide be more effective if administered post-
`prandially rather than pre-prandially to avoid potential
`development of gastroparesis and early satiety? In addi-
`tion, there were a substantial number of patients with
`Crohn’s disease enrolled in the study. In a previous study,
`teduglutide was shown to be a potentially useful therapy
`for Crohn’s disease, but only at a much greater dose
`(ⱖ0.15 mg/kg per day).35 Differential efficacy in patients
`with Crohn’s disease owing to direct effects of inflamma-
`tion on response to GLP-2, and therefore on mucosal
`repair and adaptation is, therefore, uncertain.
`The placebo response was also substantially greater in
`the current study compared with the previous study.33
`This may have been related to a less aggressive PS weaning
`protocol before randomization. The current study used an
`“optimization” period only to stabilize urine output over
`a wide range (1–2 L daily). Perhaps some patients simply
`continue their adaptation phase much longer than others,
`although only 6 patients in the teduglutide group and 8
`patients in the placebo group were ⬍2 years out (the
`previous study’s exclusion criteria) since their last bowel
`resection. We are not told whether these patients re-
`sponded differently to treatment.
`Predictors of response to teduglutide need to be either
`determined or developed. Jeppesen et al32 found that the
`length of the residual bowel was not a factor. Perhaps one
`might be a baseline meal-stimulated serum GLP-2 con-
`centration. This was unfortunately not investigated in any
`
`1418
`
`of the teduglutide or native GLP-2 studies to date despite
`the provision of standardized meals in an earlier study.34
`A study in children has identified a concentration of
`serum GLP-2, below which a need for PN can be pre-
`dicted.36 The presence of comorbid conditions that in-
`clude the health of the residual bowel and perhaps un-
`derlying pathology, mesenteric blood flow, and age are
`likely all important cofactors.
`As would be expected in the SBS/IF patient population,
`there were many adverse events in the study reported in
`this issue of GASTROENTEROLOGY, although these were
`equally distributed across
`teduglutide and placebo
`groups. Only 2 patients in the teduglutide group and 3 in
`the placebo group terminated the study owing to treat-
`ment-emergent adverse events (in both cases, abdominal
`pain that resolved within 3 days of study withdrawal in
`the teduglutide group). Abdominal pain and distention,
`nausea, peripheral edema, dyspnea, and nasopharyngitis
`were slightly more common in patients who received
`teduglutide. Stomal changes, primarily related to enlarge-
`ment, were evident in a significant minority of patients in
`the teduglutide group as would be expected, given the
`hyperplastic effect of the medication on intestinal epithe-
`lial tissue as well as previous reports.33,34 The observation
`that 1 patient may have developed a transient bowel
`obstruction during treatment with teduglutide seems in-
`conceivable to be related to the medication in the absence
`of an unrecognized pretreatment bowel obstruction given
`that teduglutide does not fertilize the growth of monster
`villi, but its use should be tempered in patients with bowel
`strictures, stomas with small lumens, or partial bowel
`obstructions.
`Concern has been raised about the potential for GLP-2
`to stimulate development of colonic adenomas in rodent
`models. The number of adenomas increased in mice
`treated with the chemical carcinogen 1,2 dimethylhydra-
`zine,37 although studies in different models, the APC-
`min/⫹ mouse, nude mice with colon cancer xenografts, or
`in GLP-2 receptor-transfected cancer cells.38 A more recent
`investigation in azoxymethane-treated mice found develop-
`ment of colonic dysplasia and adenocarcinomas in animals
`that had been chronically treated with h(Gly2)GLP-2[1-33].39
`Although the risk for malignancy is hypothetical in humans,
`and colonoscopy is difficult in these patients, colonoscopy
`should be considered at baseline for those patients with
`residual colons and perhaps even as frequently as annually
`while on therapy until more long-term safety data are
`available. This risk must be balanced against quality-of-
`life improvements, and decreased complications related to
`enhanced absorption and, therefore, portal nutrient cir-
`culation, and decreased catheter access, which may lead to
`decreased infection risk. In addition to intestine and co-
`lon, GLP-2 receptors have been found in lung, the hind-
`brain, and the hypothalamus.40 Although, to date, clini-
`
`Page 3
`
`

`
`cally detectable effects have not been observed in these
`organs, the potential exists that chronic administration of
`GLP-2 could have either beneficial or detrimental effects.
`Is teduglutide a “game changer?” Few treatments in
`SBS are. The only patients who will be able to discontinue
`PS completely will be those who are on the borderline
`between nutritional autonomy and PS dependence. Po-
`tentially, teduglutide may help some patients who sit on
`that “fence” from actually needing PS to begin with. What
`happens when teduglutide is stopped? Some preliminary
`evidence suggests the effects on adaptation may be per-
`sistent,41 although an earlier study noted histologic
`changes that trended toward baseline within 4 weeks of
`discontinuation.35 Possibly longer treatment than that
`reported in this issue of GASTROENTEROLOGY is required.42
`The advent of teduglutide, like most other new therapies,
`represents an incremental improvement in the care of
`patients with SBS/IF and likely will allow the clinician an
`additional option for patient management. Every night
`without PN is a good night, but whether teduglutide is a
`true “game changer” is not clear. Teduglutide does have
`the potential to improve quality of life for patients with
`SBS/IF, although a fully validated measure of quality of
`life in these patients awaits full development. I think we
`should look forward to the availability of teduglutide as a
`treatment for patients with SBS/IF and now we also
`eagerly await the development of longer acting analogs, as
`well as other growth factors such as HGF and KGF.
`The future is a truly artificial, or artificially grown and
`harvested, intestine; even intestinal transplantation repre-
`sents but a bridge at best. Although advances have been
`made, the practical aspects of a truly functional artificial
`gut— or even one constructed from a patient’s own stem
`cells, remains far from a clinical reality. In the meantime,
`teduglutide represents a significant, although incremental
`improvement in the treatment armamentarium for pa-
`tients with SBS/IF.
`ALAN L. BUCHMAN
`Glencoe, Illinois
`
`References
`1. Borgstrom B, Dahlqvist A, Lundh G, et al. Studies of intestinal
`digestion and absorption in the human. J Clin Invest 1957;36:
`1521–1536.
`2. Nightingale JM, Lennard-Jones JE, Walker ER, et al. Jejunal efflux
`in SBS. Lancet 1990;336-8:765–768.
`3. Buchman AL. Short bowel syndrome. In: Feldman M, Friedman LS,
`Brandt LS, eds. Sleisenger and Fordtran’s gastrointestinal and
`liver disease, 9th ed. Philadelphia: Saunders; 2010. pp. 1779 –
`1796.
`4. Buchman AL, Iyer K, Fryer J. Parenteral nutrition-associated liver
`disease and the role for isolated intestine and intestine/liver
`transplantation. Hepatology 2006;43:9 –19.
`5. Howard L, Home parenteral nutrition: survival, cost, and quality of
`life. Gastroenterology. Feb;130(2 Suppl 1):S52-S59.
`
`Editorials, continued
`
`6. Williamson RCN, Chir M. Intestinal adaptation. I. Structural, func-
`tional and cytokinetic changes. N Engl J Med 1978;298:1393–
`1402.
`7. Williamson RCN, Chir M. Intestinal adaptation. II. Mechanisms of
`control. N Engl J Med 1978;298:1444 –1450.
`8. Appleton GVN, Bristol JB, Williamson RCN. Proximal enterectomy
`provides a stronger systemic stimulus to intestinal adaptation
`than distal enterectomy. Gut 1987;28:165–168.
`intestine
`9. Solhaug JH, Tvete S. Adaptive changes in the small
`following bypass operation for obesity. Scand J Gastroenterol
`1978;13:401– 408.
`10. Levy E, Frileux P, Sandrucci S, et al. Continuous enteral nutrition
`during the early adaptive stage of the SBS. Br J Surg 1988;75:
`549 –553.
`11. Chaves M, Smith MW, Williamson RCN. Increased activity of di-
`gestive enzymes in ileal enterocytes adapting to proximal small
`bowel resection. Gut 1987;28:981–987.
`12. Gouttebel MC, Saint Aubert B, Colette C, et al. Intestinal adapta-
`tion in patients with SBS. Dig Dis Sci 1989;34:709 –715.
`13. Buchman AL, Scolapio J, Fryer J. AGA technical review on short
`bowel syndrome and intestinal transplantation. Gastroenterology
`2003;124:1111–1134.
`14. Scolapio J, Buchman AL, Floch M. Education of gastroenterology
`trainees: first annual fellows’ nutrition course. J Clin Gastroenterol
`2008;42:122–127.
`15. Messiing B, Lemann M, Landais P, et al. Prognosis of patients
`with nonmalignant chronic intestinal failure receiving long-term
`home parenteral nutrition. Gastroenterology 1995;108:1005–
`1010.
`16. Byrne TA, Wilmore DW, Iyer K, et al. Growth hormone, glutamine,
`and an optimal diet reduces parenteral nutrition in patients with
`short bowel syndrome: a prospective, randomized, placebo-con-
`trolled, double-blind clinical trial. Ann Surg 2005;242:655– 661.
`17. Johannsson G, Sverrisdottir YB, Ellegard L, et al. GH increases
`extracellular volume by stimulating sodium reabsorption in the
`distal nephron and preventing pressure natriuresis. J Clin Endo-
`crinol Metab 2002;87:1743–1749.
`18. Dworakowska D, Gueorguiev M, Kelly P, et al. Repeated colono-
`scopic screening of patients with acromegaly: 15-year experience
`identifies those at risk of new colonic neoplasia and allows for
`effective screening guidelines. Eur J Endocrinol 2010;163:21–28.
`19. Cisler JJ, Buchman AL. Intestinal adaptation in short bowel syn-
`drome. J Investig Med 2005;53:402– 413.
`20. Bulut K, Meier JJ, Ansorge N, et al. Glucagon-like peptide 2
`improves intestinal wound healing through induction of epithelial
`cell migration in vitro-evidence for a TGF-beta-mediated effect.
`Regul Pept 2004;121:137–143.
`21. Orskov C, Hartmann B, Poulsen SS, et al. GLP-2 stimulates co-
`lonic growth via KGF, released by subepithelial myofibroblasts
`with GLP-2 receptors. Regul Pept 2005;124:105–112.
`22. Dube PE, Forse CL, Bahrami J, et al. The essential role of insulin-
`like growth factor in the intestinal tropic effects of glucagon-like
`peptide-2 in mice. Gastroenterology 2006;131:589 – 605.
`23. Jeppesen PB, Hartman B, Hansen BS, et al. Impaired meal stim-
`ulated glucagon-like peptide 2 response in ileal resected short
`bowel patients with intestinal failure. Gut 1999;45:559 –563.
`24. Jeppesen PB, Hartmann B, Thulsen J, et al. Elevated plasma
`glucagon-like peptide 1 and 2 concentrations in ileum resected
`short bowel patients with a preserved colon. Gut 2000;47:370 –
`376.
`25. Dube PE, Rowland KJ, Brubaker PL. Glucagon-like peptide-2 acti-
`vates ␤-catenin signaling in the mouse intestinal crypt: role of
`insulin-like growth factor-1. Endocrinology 2008;49:291–301.
`
`1419
`
`Page 4
`
`

`
`Editorials, continued
`
`26. Bremholm L, Horum M, Henriksen BM, et al. Glucagon-like pep-
`tide-2 increases mesenteric blood flow in humans. Scand J Gas-
`troenterol 2009;44:314 –319.
`27. Brubaker PL, Drucker DJ, Sas SL, et al. Prolonged gastrointestinal
`transit in a patients with a glucagon-like peptide (GLP-1- and
`-2-producing neuroendocrine tumor. J Clin Endocrinol Metab
`2002;87:3078 –3083.
`28. Lauverjat M, Hadj AA, Vanhems P, et al. Chronic dehydration may
`impair renal function in patients with chronic intestinal failure on
`long-term parenteral nutrition. Clin Nutr 2006;25:75– 81.
`29. Jeppesen PB, Gottschalck I, Holst JJ, et al. Improvements in renal
`function in short bowel syndrome (SBS) patients treated for two
`years with subcutaneous native glucagon-like peptide 2 (GLP-2)
`[abstr] Gastroenterology 2008;134:A427–A428.
`30. Haderslev KV, Jeppesen PB, Hartmann B, et al. Short-term admin-
`istration of glucagon-like peptide-2. Effects on bone mineral den-
`sity and markers of bone turnover in short-bowel patients with no
`colon. Scand J Gastroenterol 2002;37:392–398.
`31. Buchman AL, Moukarzel A. Metabolic bone disease associated
`with parenteral nutrition. Clin Nutr 2000;19:217–231.
`32. Jeppesen PB, Pertkiewicz M, Messing B, et al. Teduglutide re-
`duces need for parenteral support among patients with short
`bowel syndrome with intestinal failure. Gastroenterology 2012;
`143:1473–1481.
`33. Jeppesen PB, Gilroy R, Pertkiewicz, et al. Randomised placebo-
`controlled trial of teduglutide in reducing parenteral nutrition
`and/or intravenous fluid requirements in patients with short bowel
`syndrome. Gut 2011;60:902–914.
`34. Jeppesen PB, Sanguinetti EL, Buchman A, et al. Teduglutide (ALX-
`0600), a dipeptidyl peptidase IV resistant glucagon-like peptide 2
`analogue, improves intestinal function in short bowel syndrome
`patients. Gut 2005;54:1224 –1231.
`35. Buchman AL, Katz S, Fang JC, et al. Teduglutide, a novel muco-
`sally active analog of glucagon-like peptide-2 (GLP-2) for the treat-
`ment of moderate to severe Crohn’s disease. Inflamm Bowel Dis
`2010;16:962–973.
`
`36. Sigalet DL, Martin G, Meddings J, et al. GLP-2 levels in infants with
`intestinal dysfunction. Pediatr Res 2004;56:371–376.
`37. Thulesen J, Hartmann B, Hare KJ, et al. Glucagon-like peptide 2
`(GLP-2) accelerates the growth of colonic neoplasms in mice. Gut
`2004;53:1145–1150.
`38. Koehler JA, Harper W, Barnard M, et al. Glucagon-like peptide-2
`does not modify the growth or survival of murine or human intes-
`tinal tumor cells. Cancer Res 2008;68:7897–7904.
`39. Lakoubov R, Lauffer LM, Trivedi S, et al. Carcinogenic effects of
`exogenous and endogenous glucagon-like peptide-2 in azoxymeth-
`ane-treated mice. Endocrinology 2009;150:4033– 4043.
`40. Yusta B, Huang L, Munroe D, et al. Enteroendocrine localization of
`GLP-2 receptor expression in humans and rodents. Gastroenter-
`ology 2000;119:744 –755.
`41. Compher C, Gilroy R, Pertkiewicz M, et al. Maintenance of paren-
`teral nutrition volume reduction, without weight loss, after stop-
`ping teduglutide in a subset of patients with short bowel syn-
`drome. J Parenteral Enteral Nutr 2011;35:603– 609.
`42. Schwartz L, Seidner D, Jeppesen PB, et al. Teduglutide for the
`treatment of short bowel syndrome-intestinal
`failure subjects
`yields further reductions in parenteral support: an interim assess-
`ment of a 2-year, open-label, phase 3 trial (STEPS2) (abstr). Am J
`Gastroenterol 2011;106:S99.
`
`Reprint requests
`Address requests for reprints to: Alan L. Buchman, MD, MSPH,
`AGAF, 959 Oak Drive, Glencoe, Illinois 60022.
`e-mail:
`.
`
`Conflicts of interest
`Dr Buchman is a former consultant for NPS pharmaceuticals.
`© 2012 by the AGA Institute
`0016-5085/$36.00
`
`Familial Visceral Myopathies: From Symptom-Based Syndromes to
`Actin-Related Diseases
`
`See “Segregation of a missense variant in en-
`teric smooth muscle actin ␥-2 with autosomal
`dominant familial visceral myopathy,” by
`Lehtonen HJ, Sipponen T, Tojkander S, et al,
`on page 1482.
`
`Mammals have genes that encode 6 different actin
`
`isoforms expressed in a developmental and tissue-
`specific fashion.1 These actins include 2 striated muscle
`(skeletal and cardiac), 2 smooth muscle (vascular and
`visceral), and 2 non-muscle actins. Isoelectic focusing sep-
`arates these actins into ␣- (skeletal, aortic smooth, and car-
`diac), ␤-, and ␥1- and ␥2- (cytoplasmic and enteric smooth
`muscle, respectively) actins. These actins share a high degree
`of structural homology, with no 2 actins differing in struc-
`ture from one another by ⬎5%. Furthermore, in mammalian
`cells, from 2 to 4 of the different actins may be simultane-
`ously present in the same cell. G-Smooth muscle actin is the
`
`1420
`
`major actin found in visceral smooth muscle, and this report
`by Lehtonen et al2 of the association of a mutation in this
`actin with familial visceral myopathy is significant for 2
`reasons. It is the first report of a human disease associated
`with this particular actin isoform and it means that now
`mutations in each of the 6 different actin isoforms have been
`shown to cause human disease.3–7 What is particularly inter-
`esting, and it is true for the mutation focused on here, is that
`in a number of cases, different diseases are caused by muta-
`tions at the same site in different actins. For example, the
`R148S mutation in ␥-smooth muscle actin causes familial
`visceral myopathy, whereas the mutation to C in ␣-smooth
`muscle actin causes thoracic aortic aneurysm and dissec-
`tion.4 The R258 mutation to either H or C in ␣-smooth
`muscle actin causes thoracic aortic aneurysm and dissec-
`tion,4 whereas in ␤-nonmuscle actin, an R to W mutation at
`the same site causes Baraitser–Winter syndrome.6 In a third
`example, a V370A mutation in ␥-nonmuscle actin causes
`autosomal-dominant nonsyndromic deafness, DFNA20/26,7
`
`Page 5

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket