Accepted Manuscript When can we cure Crohn’s? Gerassimos J. Mantzaris, MD, PhD, AGAF

PII:

S1521-6918(14)00052-3

DOI:

10.1016/j.bpg.2014.04.008

Reference:

YBEGA 1253

To appear in:

Best Practice & Research Clinical Gastroenterology

Received Date: 30 January 2014 Revised Date:

15 March 2014

Accepted Date: 13 April 2014

Please cite this article as: Mantzaris GJ, When can we cure Crohn’s?, Best Practice & Research Clinical Gastroenterology (2014), doi: 10.1016/j.bpg.2014.04.008. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

RI PT

When can we cure Crohn’s?

SC

Gerassimos J. Mantzaris, MD, PhD, AGAF

M AN U

Coordinator Consultant Gastroenterologist

Department of Gastroenterology, Evangelismos Hospital 45-47 Ypsilantou street, 10676-Athens, Greece

TE D

E-mail: [email protected]

AC C

EP

Tel. +30-213-2045223, Fax. +30-213-2041604

ACCEPTED MANUSCRIPT Abstract Crohn’s disease is a life-long idiopathic inflammatory disease which affects the entire gastrointestinal tract and occasionally extra-intestinal organs. CD is thought to result

RI PT

from complex interactions between environmental factors, the gut microbes, and the genetic background and the immune system of the host. In the last decades research on these pathogenetic components, and especially on mucosal immunity,

SC

has led to the development of biologic agents and therapeutic strategies that have improved dramatically the treatment of CD but we are still far away from curing the

M AN U

disease. If there is a treatment for CD that will probably evolve through methodical steps towards integrating research on all the components involved in the pathogenesis of CD. This holistic and global approach may aid at unraveling the mysteries of CD and developing novel agents and therapeutic strategies which by

EP

hopefully to cure.

TE D

targeting multiple pathogenetic pathways and at different stages of disease may lead

AC C

Key words: Crohn’s disease, aminosalicylates, corticosteroids, immunosuppressants, biologics, feacal microbiota transplantation, environmental factors, gut microbiota, mucosal immunity, genetics.

ACCEPTED MANUSCRIPT

Introduction The recent English edition of Wikipedia defines ‘cure’ as ‘the end of a medical

RI PT

condition; the substance or procedure that ends the medical condition, such as a medication, a surgical operation, a change in lifestyle, or even a philosophical mindset that helps end a person's sufferings’. Crohn’s disease (CD) is an idiopathic,

SC

granulomatous, life-long disease that affects any part of the gastrointestinal tract

M AN U

from the mouth to the anus and runs a relapsing-remitting or continuously active course. In up to 35% of the patients the disease affects one or more extra-intestinal organs [1]. CD can impair profoundly the quality of life and can cause significant functional and/or psychological disability [2]. Despite remarkable improvement in

TE D

the medical treatment there is still no definitive therapy for CD. Current therapeutic interventions aim at inducing and maintaining clinical and biologic remission of active disease and healing of lesions in order to restore quality of life to near normal

EP

levels and prevent complications which may lead to intestinal failure and disability [3]. Surgery is reserved for disease complications. In the text to follow, we shall

AC C

review briefly what we have learned since the first descriptions of disease, what the major obstacles to curing CD are and how these can be overcome, and, finally we will discuss whether cure is a realistic option.

A. What have we learned since the first description of CD?

ACCEPTED MANUSCRIPT Eighty years after Crohn, Ginzburg and Oppenheimer presented their landmark article [4] we have improved substantially our understanding of the pathophysiology and the natural history of CD. CD is now thought to develop in genetically susceptible individuals from complex but as yet unknown interactions between

RI PT

various environmental factors, the gut microbes (gut microbiota) and the mucosal immune system of the host [5]. These interactions determine the clinical phenotypes

in the treatment of various subtypes of CD.

SC

and the natural course of the disease. We have also made remarkable achievements

M AN U

Due to advances in molecular biology, genetics and tissue staining techniques in association with easy sampling of biologic fluids and intestinal tissues ‘bench to bedside’ research on the immune system of the host grew disproportionally faster than research on other pathogenetic components of CD. Defective innate immunity

TE D

and pathogenic effector T helper cells were considered as the driving forces for disease activation and progression. A network of cells, key-inflammatory mediators,

EP

and pro- and anti-inflammatory immune pathways were implicated in the immunopathology of CD. Defective immune regulations and apoptotic mechanisms

AC C

were incriminated for the accumulation of inflammatory cells and the perpetuation of chronic inflammation [6-10] Despite limitations, this feverish research led to the discovery and production of novel therapeutic agents, initially anti-TNFα biologics and recently anti-integrins, which target selectively key inflammatory mediators. The efficacy and safety of these agents has been elucidated in large, adequately powered randomized controlled trials (RCTs) with harder end points than simple clinical remission [11-16]. A long pipeline of non anti-TNFα biologic agents is currently being

ACCEPTED MANUSCRIPT investigated in phase II and III RCTs and is expected to further expand the pool of future therapeutic options [17-19]. Additional therapeutic interventions, such as granulocyte/monocyte apheresis [20], and autologous or allogeneic mesenchymal or hematopoietic stem cell transplantation [21-23] are also currently under

RI PT

investigation. Furthermore, pharmacokinetic studies have facilitated the initiation of therapy with appropriate doses of single drugs and/or drug combinations,

SC

monitoring the response and adherence to treatment, and optimizing therapy to ensure maximum efficacy and safety [24,25]. Modern imaging modalities allow

M AN U

better localization and grading of disease activity and severity. Factors predicting a disabling course of CD have been identified and call upon earlier therapeutic interventions to prevent irreversible bowel damage. Thus, we are now able to deliver highly individualized therapies based not only on disease profile but also on

TE D

efficacy and safety of medications and on important elements of patient profiles. A ‘treat-to-target’ strategy is evolving which marries the concepts of early intervention and regular assessment of disease activity to ensure optimal control of disease using

EP

objective serological and faecal markers of inflammation and mucosal healing as

AC C

surrogate markers of sustained ‘deep’ remission of CD [3]. This strategy promises excellent mid-term therapeutic outcomes, including reduced rates of hospitalizations and surgery, improvement in health-related quality of life and avoidance of disability.

Intensive research has documented the important role of each of these pathogenetic components of CD. Epidemiological studies have identified amongst others particular risk factors for developing and a disabling course of disease; geographical

ACCEPTED MANUSCRIPT gradients in the prevalence of disease; and, a dramatic increase in the incidence when former underprivileged societies adopt western dietary and life-styles and improve hygienic conditions [1,26]. A ‘clean-environment’, linked to the hygiene hypothesis, elimination of helminthes, extensive and repetitive use of antibiotics in

RI PT

early life, infections, smoking, appendectomy, and dietary modifications both qualitative and quantitatively have all been implicated with various levels of

SC

evidence in disease pathogenesis [1,5-7].

Our knowledge on the genetic variants of CD has truly exploded in the last decades.

M AN U

We now know that CD is a multi-genic disease; different gene variants control innate and adaptive immunity. At least in Caucasians, specific genes of the innate immunity, such as the NOD2/CARD15, are implicated in the pathogenesis of CD [27]. Childonset CD has a stronger genetic predisposition compared to adult-onset CD [28].

TE D

Despite genetic diversity, universally distinct clinical phenotypes have been identified which may also be influenced by genetic variants [29] and have led to a

EP

classification of CD based on age of onset, anatomic distribution, clinical behaviour, and involvement of the perianal area; this classification has important prognostic

AC C

and therapeutic implications [30]. The long neglected human microbiota is now attracting the interest of researchers after recent revolutionary molecular biology techniques and compositional sequence technology have enabled the culture of mixed populations of gut microbes and the genetic composition of the microbiota (microbiome)[31-33]. The gut microbiota, a huge body in the human body, comprises by trillions of microbes most of which astonishing belong only to four phyla, predominantly Firmicutes and Bacteroidetes

ACCEPTED MANUSCRIPT and to a lesser degree Proteobacteria and Actinobacteria [33]. The microbiota develops early during childhood under the influence of genetic, dietary and life-style factors, ethnicity, and geography and then remains stable throughout the adult [3436]. In adulthood, the composition and function of the microbiota may be affected

RI PT

by dietary factors, especially prebiotics (oligo- and polysaccharides), antibiotics, infections, smoking and appendectomy [34,35,37]. The gut microbes are in intimate

SC

and dynamic coexistence with the gut epithelium which allows selective absorption of nutrients, the regulation of fat storage [38], vitamin production, and the

probably under genetic [40].

M AN U

maturation and function of the immune system [6,38,39]. This ‘cross-talking’ is

The unequivocal role of the microbiota in the development of intestinal inflammation in CD has been demonstrated in animal models, improvement of

TE D

colonic involvement after faecal diversion followed by relapse when ileostomy effluents are infused into the diverted colon or by infusion of intestinal contents in

EP

the excluded ileum, the post-operative recurrence and the effect of antibiotics for post-operative prophylaxis [26,41-43]. In CD, there is reduced diversity and altered

AC C

function of the microbiota (dysbiosis) [35,44-46]. Numbers of firmicutes, lactobacilli, bifidobacteria, and bacteroidetes decrease whereas Faecalibacterium prausnitzii, an anti-inflammatory commensal which consists 5% of the normal microbiota, is reduced [47,48]. In contrast, number of facultative bacteria, proteobacteria and adherent-invasive Escherichia coli increase. The composition of the microbiota in relatives of patients with CD is more similar to patients than healthy individuals [49].

ACCEPTED MANUSCRIPT Metagenomics have shown that dysbiosis alters fundamental metabolic functions of the microbiota [37]. Finally, many surgical dogmas have been challenged in the last 35 years. Indications

RI PT

and timing for surgical intervention have been revised, surgical techniques have improved, new techniques have been introduced, and the number of operations has probably declined. Complicated rather than medically intractable disease is now the

SC

main drive for surgical therapy; strictureplasty has replaced intestinal resection for short intestinal strictures; examination under anaesthesia, abscess drainage, and

M AN U

seton placement under pelvic MRI guidance has become the first intervention in a stepwise multidisciplinary approach to complex perianal fistulae; and, early surgery may be a valid option for patients with limited stricturing ileocolonic disease [26,28]. Meta-analyses of RCTs and non-RCTs have shown that laparoscopic surgery is equally

TE D

safe to the open operation but decreases the rates of perioperative complications

EP

and incisional hernia [50].

AC C

B. Why have we not achieved ‘cure’ of CD?

Despite great achievements in the pathophysiology and treatment important questions relevant to ‘curing’ CD remain unanswered: what causes the disease, why is innate immune defective, what drives flares and spontaneous remission, how the microbiota influences immune regulation, why the disease is discontinuously and unevenly distributed in the gut, why a proportion of patients develop perianal

ACCEPTED MANUSCRIPT disease or extra-intestinal manifestations, etc. Current medical therapy can ensure at best a mid-term ‘honeymoon’ rather than ‘cure’. Dietary modifications, exclusive elemental nutrition, modifiers of the microbiota (antibiotics, probiotics, prebiotics, synbiotics, faecal microbiota transplantation) cannot ‘cure’ the disease [26,28,51-

RI PT

59]. Corticosteroids can induce clinical remission in a proportion of patients but cannot effectively heal lesions and are associated with serious adverse events.

SC

Remission rates on thiopurines fall as time goes by and disease will relapse if treatment is withdrawn [26,60,61]. Approximately one fourth of patients will be

M AN U

primary non responders to anti-TNFα agents and another third may lose response overtime or develop serious allergic or infectious complications [26]. Co-morbidities may not allow administration of the ‘best’ medical therapy, especially in aged patients. Biosimilars can only promise access of a proportion of underprivileged

TE D

patients to biologics. Based on available data, non-anti-TNFα’ biologics may be helpful for patients with disease refractory to prior therapies but do not seem to offer ‘cure’. Failure of current medical therapy is best underscored by lack of

EP

adherence, and use of therapies of questionable efficacy and safety. Interestingly,

AC C

approximately one fifth of patients in RCTs will respond or achieve remission on placebo [62-64] and a small group of patients in the community may not need any treatment [26]. Last but not least, despite ‘curative’ surgical resection and postoperative prophylaxis CD will inevitably recur [28]. Faecal diversion may alleviate only temporarily symptoms of colitis and complicated perianal disease. Yet, CD remains an incurable disease. Why?

ACCEPTED MANUSCRIPT • Progress in medicine is slow. A characteristic example is the discovery of H. pylori, a simple infectious agent as a cause of peptic ulcer disease, a plague of mankind for thousands of years and a monumental failure of epidemiologists, biologists

RI PT

and pathologists.

• Medical education probably no longer produces this species, ‘researcher-

SC

physicians’. In many countries, medical students are exposed to cataclysmic information during a rather strictly specialty-orientated training rather than

M AN U

proper medical education that would aim at creating personalities with restless minds and influences from various medical and nonmedical disciplines. Separation created a chasm in communication and cooperation between basic scientists and clinicians which has resulted in partition and isolation of the

TE D

medical disciplines and inevitably to ‘paucity of convergent thinking’ or luck of

EP

‘thinking across the boundaries of traditional disciplines’ [39].

• Universal lack of adequate funding and strong competition for the limited

AC C

resources has further intensified fragmentation and privacy in the medical science.

• Implementation of market rules and practices in the Academic Society in association with the evaluation of the scientific work on the basis of publication records and impact factors (‘publish or perish’) is leading young investigators to

ACCEPTED MANUSCRIPT massive research of questionable quality leaving aside the innovative and pioneering research. All these are not applicable exclusively to IBD. In addition, what we have witnessed

RI PT

in CD is an isolated and disproportionally allocated research mainly on those components of disease pathogenesis that are especially related to the host. However, as Fiocchi points out in a recent excellent editorial, the isolated

SC

modulation of each of the pathogenetic components of CD is unlikely to lead to ‘cure’ [65]. Thus,

M AN U

• for many years, epidemiological studies have documented strong relations between various environmental factors and CD but have failed to correlate aetiopathogenetically any of these factors to mechanisms underlying and/or

TE D

potentially implicated in the pathogenesis of this disease [39].

• the human genome has not changed in the last 100 years! Although ‘cases’ of

EP

intestinal involvement resembling CD were described centuries ago, CD as is

AC C

known today was described convincingly nearly 100 years ago. Genetics studies have revealed an astonishing number of genetic loci associated with CD but these associations can explain only a limited proportion of cases. CD is a multi-genic disease and no single genetic association can explain its diverse clinical phenotypes. Recent experimental work has shown that manipulating some important genetic variants, such as NOD2, may alleviate symptoms of chemicallyinduced colitis [66] but in humans none of these associations can offer

ACCEPTED MANUSCRIPT convincingly solid ground for a practical approach to patient treatment let alone ‘cure’ of CD.

• although there are numerous animal models of CD, none of these addresses the

RI PT

complexity of the interactions between the microbiota, the immune system and the environmental factors in CD. This is evidenced by disparity in the results of

M AN U

their efficacy to treat acute experimental colitis.

SC

RCTs and meta-analyses of probiotics in the treatment of human CD [26,56] and

• the limitations of treatments aiming at resetting disordered mucosal immune mechanisms have been underscored earlier. The activation of the mucosal immune system in CD resembles sequential explosions (activation of immune

TE D

pathways) after firing a single firework (the initial trigger). Targeting each individual immune pathway, especially those that are activated late in the cascade of immune events may control to a certain degree the inflammatory

EP

response but cannot cure CD. Blocking some immune pathways with biologics

AC C

may lead onto paradoxical inflammation which may be more severe that CD itself [7,68]

• manipulating the microbiota has proven efficacious in the prevention and treatment of several conditions, such as pouchitis, hepatic encephalopathy, Listeria infections and travelers’ diarrhoea. In a mouse model of colitis administration of F. Prausnitzii strain A2-165 protected against 2,4,6trinitrobenzenesulfonic acid (TNBS)-induced colitis [48]. However, CD is a much

ACCEPTED MANUSCRIPT more complex disease characterized by severe dysbiosis. Hopes that empirical treatment with a single probiotic or combinations of probiotics may restore a

C. What should be done to achieve ‘cure’ of CD?

RI PT

healthy microbiota and ‘cure’ CD is in itself a childish naivety.

SC

• Great achievements in Medicine came from people who had global influences from various disciplines and had time to think in a simple way. We must rethink

M AN U

CD! Why the disease was unknown just a century ago? What has truly changed in our internal and external environment that has led to the appearance of this horrible disease? Why the disease has a predilection for Caucasians and particular ethnicities? What drives the dramatic increase in prevalence in societies that are

TE D

rapidly westernized? Is the inverse epidemiological association of tuberculosis to CD simply the result of improved hygienic conditions or they are connected

EP

aetiologically? Is CD truly a multifactorial disease or what we see at the time of diagnosis is a complex phenotype that is being built gradually on a background

AC C

platform set by a single infectious agent?

• These simple questions make crystal clear that we see the tree and miss the forest. We are lost in the curling corridors of the labyrinth of research that is compartmentalized in narrow, isolated, and strictly demarcated scientific fields without an Ariadne’s thread! This myopic approach has resulted in the accumulation of a huge load of information on each of the pathogenetic

ACCEPTED MANUSCRIPT components of CD, especially mucosal immunity, but not to global knowledge that can be transformed into revolutionary research towards a cure of CD. We need to ‘U-turn’, rethink, design and execute cooperative, integrated research that will include all the pathogenetic components of CD [65]. Hopefully, we have

RI PT

recently begun to witness research moving towards this direction [37].

SC

• An important first step could be to orchestrate national and international interdisciplinary consortiums of scientists from medical and nonmedical

M AN U

disciplines. These will have the difficult task to review what is known, identify missing links between the factors associated with the pathogenesis of CD, and then prioritize the next essential steps of innovative, pioneering integrated research which would address the unanswered questions in the pathogenesis and

TE D

treatment of CD. Focusing on societies which undergo rapid and dynamic environmental changes as well as on early childhood during crucial changes in the composition of the microbiota and maturation of immune system rather than late

EP

adulthood may be more appropriate targets.

AC C

• Research on the human microbiota should drive this integrated research. The microbiota may be the ‘missing link’ between external environmental factors, the genetic background and the mucosal immune system of the host. The interplay between external environmental factors and dysbiosis in genetically predisposed subjects may initiate immune reactions that lead onto gut damage. Indeed, environmental factors, especially diet and antibiotics, may cause dysbiosis during early, critical developmental stages of the immune system. Metagenomis have

ACCEPTED MANUSCRIPT shown that dysbiosis alters fundamental metabolic functions of the microbiota [37]: breaks down the gut barriers and impacts severely on the host immune response to microbes by inducing activation of effector T helper cells or suppressive T regulatory cells, the IL-23/Th1/Th17 pathway, and autophagy

RI PT

[69,70]. Dysbiosis-induced impairment in the enzymatic activity of the microbiota modifies the composition of luminal bile acids which may erase the anti-

SC

inflammatory effects of some bile acids contributing to chronic inflammation in IBD [71]. Noteworthy, mutant genes implicated in the pathogenesis of CD can

M AN U

regulate the host immune response to environmental factors (e.g. autophagy genes) and code for sensors of intestinal bacteria (e.g. NOD2) [6,39,72]. Recent experimental data suggests that NOD2 deficiency causes dysbiosis which renders the colonic epithelium vulnerable to chemical injury and colitis [73].

TE D

Consequently, disease-related dysbiosis can be manipulated by targeting the NOD2 gene [66]. Manipulation of these pathogenetic pillars of CD is more likely to work in child-onset CD, during crucial changes in the composition of the

EP

microbiota and maturation of immune system. Furthermore, human microbiota-

AC C

driven excess fat storage may trigger pro-inflammatory immune effects offering a theoretical bridge between improved socio-economic conditions, altered dietary habits and rapid changes in the epidemiology of CD [39].

• The human microbiota may be used for mining new drugs and identify biomarkers to monitoring the effect of treatment on the course of disease [37,39,45]. These biomarkers are expected to be of higher specificity and sensitivity than traditional serological or faecal biomarkers.

ACCEPTED MANUSCRIPT

• Furthermore, the effect of current medical treatments on the composition and function of the gut microniota and the secondary effects of this particular dysbiosis on the response to treatment and the course of CD should be

RI PT

thoroughly investigated longitudinally. There is preliminary evidence that treatment-induced disordered composition and function of the microbiota may

SC

affect metabolism and nutrient availability in the presence of oxidative stress and tissue injury [45]. Exploring these interactions may help in explaining primary non

M AN U

response or secondary loss of response to medications and treatment-induced paradoxical inflammation, and may select patients for treatment optimization or switch to alternative therapies.

TE D

• Research should expand to incorporate additional fields. For instance, the function of the mucosal immune system is not controlled exclusively by environmental factors, the microbiota and genetic variants of the host but also by

EP

neurophysiological and neuroendocrine mechanisms. The role of neural

AC C

regulation of the immune system in various intestinal diseases, especially irritable bowel syndrome, has been recognized for many years and vagal-induced/cytokine mediated inhibition of active intestinal inflammation is a known homeostatic mechanism. Although elements of psychological, social and physical stress have been incorporated in measurements of quality of life in CD and anti-depressants consist often an integral part of the therapy research probably underestimates the importance of the brain-gut axis in IBD.

ACCEPTED MANUSCRIPT • Unfortunately, pioneer research is nowadays very costly and cannot be adequately funded by independent sources. Funding for clinical research comes mainly from the Pharmaceutical Industry. However, the priorities of the Industry cannot usually meet the needs of pure Science. Industry initiated trials are

RI PT

subjected to market forces and barriers from Regulatory Authorities. For instance, a recent publication claimed that many ‘real-life’ patients would never qualify for

SC

inclusion in any trial of anti-TNFα biologics [74]. As it is likely that in foreseeable future the industry will continue to invest on novel agents that modulate immune

M AN U

pathways hoping for fast rewards we should urgently strive for independent studies targeting unmet needs in CD pathogenesis and therapy.

• Funding should originate from independent sources: scientific, governmental or

TE D

international organizations, patient associations, but also where possible by the corporate members of scientific organizations. Research protocols that involve all the pathogenetic components of CD should be preferentially funded. Research

EP

products should be accessible to enhance productive criticism and creative

AC C

brainstorming.

D. Is cure of Crohn’s disease a realistic option?

To ‘cure’ CD we must learn from and correct our mistakes. Instead of accumulating irrelevant and redundant data on each individual pathogenetic component following the trends of time we should orchestrate globally integrated research to study

ACCEPTED MANUSCRIPT concurrently the functional interplay between environmental factors, the dysbiotic microbiota, the mucosal immune system, the genetic background of the host and the neuroendocrine control of the gut function taking advantage of recent revolutionary technological advances, such as proteomics, metagenomics, transcriptomics,

RI PT

metabolomics, epigenetics, etc. The effect of any acquired disorder affecting each of these components, for instance the effect of smoking or prolonged antibiotic use on

SC

the microbiota, should be longitudinally studied to assess its impact(s) on the other pathogenetic components. Mining novel and disease specific biomarkers from the

M AN U

dysbiotic microbiota will aid at following up the course of disease and linking these effects to measurable outcomes of disease. In this way, we will probably be able to conceive new ideas, produce novel therapeutic agents, and apply novel therapeutic strategies addressing multiple pathogenetic pathways which may be used either to

TE D

prevent or to ‘cure’ CD.

Identifying novel predictors of future disease, genetic, immune, environmental,

EP

microbiota-derived may help in selecting patients at a pre-clinical stage for

AC C

appropriate preventive interventions. One such approach in off-springs of affected families may be to select those with established susceptibility genes and manipulate any culprit environmental factor(s) during the early, crucial developmental stages of the microbiota or even colonize the offspring using faecal microbiota transplantation from unaffected individuals to prevent dysbiosis-initiated immune events leading onto CD. Identifying novel microbiota-derived antigens from patients with CD, especially affected family members, may also open another avenue of disease prevention or treatment using oral immunization as we have done in the past for

ACCEPTED MANUSCRIPT orally transmitted intestinal and systemic infections. These therapeutic strategies may also apply for patients who have undergone ‘therapeutic’ resection for irreversible bowel damage.

RI PT

CD is not a single entity but consists of various subsets/clinical phenotypes (Crohn’s diseases) with unpredictable clinical course and response to treatment. If cure is ‘a

SC

substance or procedure that ends a medical condition’ it is probably naïve to believe that in the foreseeable future ‘cure’ of CD will be a Procroustes bed, an ‘one-fit-all’

M AN U

single regimen that can apply to all patients independently of the stage of disease and the accumulated intestinal damage. Therefore, timing of intervention may continue to be crucial. This is more important for childhood CD because children will live with the disease for longer periods and avoidance of surgeries and disability is

TE D

much more important than in adulthood CD [75]. Using a variety of combination therapies we could target in a highly individualized way the particular clinical phenotype at different stages of the evolution of CD in the particular patient. Thus,

EP

patients with established disease may be treated by blocking all the involved

AC C

pathogenetic components and prolonged follow up using microbiota-derived biomarkers may ensure ‘cure’. For instance, attacking crucial genetic variants linked to CD, such as NOD2, in association with dietary modifications, immune modulation and manipulation of the microbiota [66]. Alternatively, production of fibrolytic agents may substitute for surgical strictureplasty or resection of stenotic bowel areas and decrease the need for surgical intervention in patients with irreversible bowel damage.

ACCEPTED MANUSCRIPT Any ‘realistic’ option assumes that is potentially valid, has been adopted as working hypothesis by the international scientific society, and is generously financed. How long it will take such an effort is purely conjectural. The ‘Craft is long’ but although the pace of any such innovative research effort is unpredictable, random events as in

RI PT

the H. pylori story, may accelerate tremendously the sequence of events towards ‘seeing light at the end of the tunnel’ as long as we are keeping the train on the

SC

tracks.

M AN U

We should be dreamers but we must also be realistic. Currently, we cannot give our patients their lives back. However, until the mysteries of CD are unraveled and because a universal lack of quality of care in IBD has been documented [76,77], every effort should be made to offer patients a safe, timely administered, effective,

TE D

evidence-based, and patient-centered healthcare [78]. Amongst others, this includes universally agreed structure, process and outcome measures based on solid quality indicators; expansion of centers of excellent care for easy access of patients to

EP

multidisciplinary expert teams and regular audits for continuous improvement;

AC C

allocation of sufficient time in motivational interviews to express expectations of life and concerns for their disease and treatments; interactive education for groups of patients with similar disease burden rather than incomprehensible leaflets. Furthermore, medical therapy should be personalized and administered by better drug delivery systems as early as possible to prevent irreversible gut damage. Treatment should aim beyond symptomatic control to include normalization of markers of inflammation, mucosal healing and, if possible, histologic remission. Combination therapies targeting different inflammatory pathways may achieve

ACCEPTED MANUSCRIPT tighter control of disease. Patients should be monitored regularly so that treatment is adapted to the state of disease. These mid-term outcome measures are expected to relieve the physical and psychological burden of CD and allow patients to run a

RI PT

normal life under a safe treatment until definitive therapy is found. Summary

For many years medical treatment of CD was empiric and surgery was reserved for

SC

incapacitating symptoms and complications. After the 1980s surgery moved to the

M AN U

backstage; corticosteroids became the mainstay of therapy for active disease and thiopurines the drugs of choice for remission maintenance and post-operative prophylaxis. The advent of anti-TNF was a breakthrough in the management of patients with disease refractory to prior treatments. Advanced technology, novel

TE D

markers of inflammation, pharmacokinetic and genetic studies have allowed better localization of disease and close monitoring of response to treatment over time. Non-anti-TNFα biologics are under investigation and approval of safer anti-integrins

EP

is pending. As for rheumatoid arthritis, a ‘teat-to-target’ strategy is evolving for CD and promises better long-term outcomes through tighter control of inflammation

AC C

and mucosal healing by continuous fine adjustments of treatment. However, CD remains incurable. This is probably because research has focused exclusively on the host and on isolated pathogenetic components of disease. Recent advances have shifted our interest to the lumen: the gut microbiota, a huge organ within the human body, co-existing and continuously interacting with the immune system and the genetic constituents of the host, emerges as a new therapeutic target, a source for novel biomarkers, and a goldmine for new drug development. However, focusing

ACCEPTED MANUSCRIPT only on the microbiota will undoubtedly lead to the same mistakes, i.e. accumulating information on another pathogenetic pillar. Using effectively the available resources consortiums of basic and clinical medical and nonmedical scientists should prioritize and execute integrated ‘holisitc’ research to include all factors that are relevant to

RI PT

the pathogenesis of CD. This global approach may lead to the development of novel agents and therapeutic strategies destined to ‘cure’ CD. However, until the mysteries

SC

of CD are uncovered every effort should be made to ensure that excellent patientcentered healthcare is delivered in appropriately organized Centers of Excellent Care

M AN U

by multidisciplinary expert teams.

Practice points

Crohn’s disease is a life-long idiopathic inflammatory bowel disease



There is no definitive medical or surgical therapy.



Current medical therapy aims at inducing ‘deep’ clinical, biologic and

TE D



EP

endoscopic remission; deep remission is associated with reduced rates of

AC C

complications, hospitalization and surgeries at least in the short-term. •

The long-term efficacy of a ‘treat-to-target’ therapeutic strategy needs to be investigated in prospective controlled trials.



Until CD is cured every effort should be undertaken to ensure the delivery of personalized, holistic, effective, and safe therapy to patients by multidisciplinary teams of experts in centers of excellent care.

ACCEPTED MANUSCRIPT •

Although structure and process measures are central to the delivery of optimal treatment, universally applied outcome measures is all that matters

RI PT

to patients and need to be defined.

Research agenda •

Epidemiological and genetic studies and research focusing on the host have

SC

been unable to discover what causes the disease and lead to ‘cure’.

The gut microbiota emerges as a novel field for innovative research.



The interactions between constituents of the gut microbiota, dietary factors,

M AN U



genes, and the mucosal immune system of the host need to be defined. •

Research should also assess whether the microbiota can be used for mining



TE D

new drugs and/or as a biomarker to monitoring the effect of treatment. However, continuing isolated research on each of the pathogenetic components of CD in our struggle to identify the aetiopathogenesis of CD is



EP

expected to be counter-productive. Consortiums of scientists from different disciplines should be involved in

AC C

innovative integrated research focusing on the functional interplay between the components involved in the pathogenesis of CD.



Early age and societies undergoing rapid environmental and socioeconomic changes should be the focus of this research.

ACCEPTED MANUSCRIPT References

[1]. Langholz E. Current trends in inflammatory bowel disease: the natural history.

RI PT

Therap Adv Gastroenterol 2010;3:77-86. [2]. Peyrin-Biroulet L, Cieza A, Sandborn WJ et al. Development of the first disability index for inflammatory bowel disease based on the international classification of

SC

functioning, disability and health. Gut 2012;61:241-247.

M AN U

[3]. Bouguen G, Levesque BG, Feagan BG et al. Treat to Target: A Proposed New Paradigm for the Management of Crohn's Disease. Clin Gastroenterol Hepatol ‘(In press)’.

*[4]. Crohn BB, Ginzburg L & Oppenheimer GD. Regional ileitis: A pathologic and

TE D

clinical entity. 1932. Mt Sinai J Med 2000;67:263-268. [5]. Van Assche G, Dignass A, Panes J et al for the European Crohn's and Colitis

EP

Organisation (ECCO). The second European evidence-based consensus on the diagnosis and management of Crohn's disease: Definitions and diagnosis. J Crohn's

AC C

Colitis 2010;4:7-27.

[6]. Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol 2008;8:458-466. [7]. Steinhart AH, Tolomiczenko G. IBD 2009: Emerging Research Frontiers on the path to a cure. Can J Gastroenterol 2010;24:557-565.

ACCEPTED MANUSCRIPT [8]. Gersemann M, Wehkamp J, Stange EF. Innate immune dysfunction in inflammatory bowel disease. J Intern Med 2012; 271: 421-428. [9]. Marks DJ, Harbord MW, MacAllister R et al. Defective acute inflammation in

RI PT

Crohn’s disease: a clinical investigation. Lancet 2006;367:668-678. [10]. Brand S. Crohn’s disease: Th1, Th17 or both? The change of a paradigm: new

Crohn’s disease. Gut 2009;58:1152-1167.

SC

immunological and genetic insights implicate Th17 cells in the pathogenesis of

M AN U

*[11]. Targan SR, Hanauer SB, van Deventer SJ et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. N Engl J Med 1997;337:1029-1035.

[12]. Hanauer SB, Feagan BG, Lichtenstein GR et al. Maintenance infliximab for

TE D

Crohn’s disease: the ACCENT 1 randomised trial. Lancet 2002;359:1541-1549. [13]. Present DH, Rutgeerts P, Targan S et al. Infliximab for the treatment of fistulas

EP

in patients with Crohn’s disease. N Engl J Med 1999;340:1398-1405.

AC C

[14]. Sands BE, Anderson FH, Bernstein CN et al. Infliximab maintenance therapy for fistulizing Crohn's disease. N Engl J Med 2004;350:876-885. [15]. Hanauer SB, Sandborn WJ, Rutgeerts P et al. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology 2006;130:323-333.

ACCEPTED MANUSCRIPT [16]. Colombel JF, Sandborn WJ, Rutgeerts P et al. Adalimumab for maintenance of clinical response and remission in patients with Crohn's disease: the CHARM trial. Gastroenterology 2007;132:52-65.

RI PT

[17]. Sandborn WJ, Feagan BG, Fedorak RN et al. A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-tosevere Crohn's disease. Gastroenterology 2008;135:1130-1141.

SC

[18]. Sandborn WJ, Gasink C, Gao LL et al. Ustekinumab induction and maintenance

M AN U

therapy in refractory Crohn's disease. N Engl J Med 2012;367:1519-1528. [19]. Sandborn WJ, Feagan BG, Rutgeerts P et al. Vedolizumab as induction and maintenance therapy for Crohn’s disease. N Engl J Med 2013;369:711-721. [20]. Sands BE, Katz S, Wolf DC et al. A randomised, double-blind, sham-controlled

TE D

study of granulocyte/monocyte apheresis for moderate to severe Crohn's disease. Gut 2013;62:1288-1294.

EP

[21]. van Deen WK, Oikonomopoulos A & Hommes DW. Stem cell therapy in inflammatory bowel disease: which, when and how? Curr Opin Gastroenterol

AC C

2013;29:384-390.

[22]. Voswinkel J, Francois S, Simon JM et al. Use of mesenchymal stem cells (MSC) in chronic inflammatory fistulizing and fibrotic diseases: a comprehensive review. Clin Rev Allergy Immunol 2013;45:180-192. [23]. Hawkey CJ. Stem cells as treatment in inflammatory bowel disease. Dig Dis 2012;30 Suppl 3:134-139.

ACCEPTED MANUSCRIPT

[24]. Hindorf U, Lyrenäs E, Nilsson A & Schmiegelow K. Monitoring of long-term thiopurine therapy among adults with inflammatory bowel disease. Scand J

RI PT

Gastroenterol 2004;39:1105-1112. [25]. Steenholdt C, Brynskov J, Thomsen OO et al. Individualized therapy is more cost-effective than dose intensification in patients with Crohn's disease who lose

SC

response to anti-TNF treatment: a randomized, controlled trial. Gut ‘(in press)’.

M AN U

[26]. Dignass A, Van Assche G, Lindsay JO et al. The second European evidence-based consensus on the diagnosis and management of Crohn's disease: current management. J Crohn's Colitis 2010;4:28-62.

*[27]. Hugot JP. CARD15/NOD2 mutations in Crohn’s disease. Ann N Y Acad Sci

TE D

2006;1072:9-18.

[28]. Van Assche G, Dignass A, Reinisch W et al. The second European evidence-

EP

based consensus on the diagnosis and management of Crohn's disease: special situations. J Crohn's Colitis 2010;4:63-101.

AC C

[29]. Cleynen I, González JR, Figueroa C et al. Genetic factors conferring an increased susceptibility to develop Crohn’s disease also influence disease phenotype: results from the IBDchip European Project. Gut 2013;62:1556–1565. [30]. Satsangi J, Silverberg MS, Vermeire S & Colombel JF. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut 2006;55:749-753.

ACCEPTED MANUSCRIPT *[31]. Turnbaugh PJ, Ley RE, Hamady M et al. The human microbiome project. Nature 2007;449:804–810. [32]. Goodman AL, Kallstrom G, Faith JJ et al. Extensive personal human gut

Proc Natl Acad Sci USA 2011;108:6252-6257.

RI PT

microbiota culture collections characterized and manipulated in gnotobiotic mice.

[33]. Structure, function and diversity of the healthy human microbiome. Nature

SC

2012;486:207-214.

M AN U

[34]. Palmer C, Bik EM, DiGiulio DB et al. Development of the human infant intestinal microbiota. PLoS Biol 2007;5:1556-1573.

[35]. Prideaux L, Kang S, Wagner J et al. Impact of ethnicity, geography, and disease

2013;19:2906-2918.

TE D

on the microbiota in health and inflammatory bowel disease. Inflamm Bowel Dis

[36]. Bernstein C. Assessing environmental risk factors affecting the inflammatory

EP

bowel diseases: A joint workshop of the Crohn’s disease-40 years on. Br Med J

AC C

1978;2:1106-1107.

*[37] Juste C, Kreil DP, Beauvallet C et al. Bacterial protein signals are associated with Crohn's disease. Gut 2014; in press. *[38]. Backhed F , Ding H , Wang T et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 2004;101:15718–15723. [39]. Shanahan F. Gut microbes: from bugs to drugs. Am J Gastroenterol 2010;105:275-279.

ACCEPTED MANUSCRIPT [40]. Cerf-Bensussan N, Gaboriau-Routhiau V. The immune system and the gut microbiota: friends or foes? Nat Rev Immunol 2010;10:735–744. [41]. Rath HC, Herfarth HH, Ikeda JS et al. Normal luminal bacteria, especially

RI PT

Bacteroides species, mediate chronic colitis, gastritis, and arthritius in HLAB27/human β2 microglobulin transgenic rats. J Clin Invest 1996;98:945–953.

[42]. D’Haens GR, Geboes K, Peeters M et al. Early lesions of recurrent Crohn’s

SC

disease caused by infusion of intestinal contents in excluded ileum. Gastroenterology

M AN U

1998;114:262–267.

[43]. Rutgeerts P, Hiele M, Geboes K et al. Controlled trial of metronidazole treatment

for

prevention

of

Crohn’s

recurrence

after

ileal

resection.

Gastroenterology 1995;108:1617–1621.

TE D

*[44]. Seksik P, Rigottier-Gois L, Gramet G et al. Alterations of the dominant faecal bacterial groups in patients with Crohn’s disease of the colon. Gut 2003;52:237–242.

EP

[45]. Morgan XC, Tickle TL, Sokol H et al. Dysfunction of the intestinal microbiome in

AC C

inflammatory bowel disease and treatment. Genome Biology 2012;13:R79. [46]. Manichanh C, Rigottier-Gois L, Bonnaud E et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006;55:205–211.

[47]. Sokol H, Piqneur B, Watterlot L et al. Faecalibacterium prouznitzii is an antiinflammatory commensal bacterium identified by gut microbiota analysis of Crohn’s disease patients. Proc Natl Aced Sci USA 2008;105:16731-16736.

ACCEPTED MANUSCRIPT [48]. Miquel S, Martín R, Rossi O et al. Faecalibacterium prausnitzii and human intestinal health. Curr Opin Microbiol 2013;16:255-261. [49]. Hedin C, McCarthy NE, Louis P et al. Siblings of patients with Crohn's disease

RI PT

have a distinct microbiological and immune phenotype compared to healthy controls: insights into disease pathogenesis. Gastroenterology 2013;144:Suppl 1:S57–58.

SC

[50]. Patel SV, Patel SVB, Ramagopalan SV & Ott MC. Laparoscopic surgery for

M AN U

Crohn’s Disease: a meta-analysis of perioperative complications and long term outcomes compared with open surgery. BMC Surgery 2013; 13: 14. [51]. Mendizabal M, Blonski W, Kotlyar D & Licthenstein GR. Antibiotic use in the treatment of Crohn’s disease. In Lichtenstein GR (ed) Crohn’s disease: the complete

TE D

guide to medical treatment, pp 119-133. New Jersey: Slack Incorporated, 2011. [52]. Griffiths AM, Ohlsson A, Sherman PM et al. Meta-analysis of enteral nutrition as

1067.

EP

a primary treatment of active Crohn’s disease. Gastroenterology 1995;108:1056-

AC C

[53]. Richman E & Rhodes JM. Review article: evidence-based dietary advice for patients with inflammatory bowel disease. Aliment Pharmacol Ther 2013;38:11561171.

[54]. Talley NJ, Abreu MT, Achkar JP et al. An evidence-based systematic review on medical therapies for Inflammatory Bowel Disease. Am J Gastroenterol 2011;106:S2– S25.

ACCEPTED MANUSCRIPT [55]. Khan KJ, Ullman TA, Ford AC et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol. 2011;106:661673.

RI PT

[56]. Doherty GA, Bennett GC, Cheifetz AS & Moss AC. Meta-analysis: targeting the intestinal microbiota in prophylaxis for post-operative Crohn's disease. Aliment Pharmacol Ther 2010;31:802-809.

SC

[57]. Duricova D, Pedersen N, Elkjaer M, Jensen JK & Munkholm P. 5-aminosalicylic

Colitis 2010;4:575-581.

M AN U

acid dependency in Crohn's disease: a Danish Crohn Colitis Database study. J Crohn’s

[58]. Feagan BG, Sandborn WJ, Mittmann U et al. Omega-3 free fatty acids for the maintenance of remission in Crohn disease: the EPIC randomized controlled trials.

TE D

JAMA 2008;299:1690–1697.

[59]. Turner D, Zlotkin SH, Shah PS & Griffiths AM. Omega 3 fatty acids (fish oil) for

EP

maintenance of remission in Crohn's disease. Cochrane Database Syst Rev 2009;1:CD006320.

AC C

[60]. Talley NJ, Abreu MT, Achkar JP et al. An evidence-based systematic review on medical therapies for Inflammatory Bowel Disease. Am J Gastroenterol 2011;106:S2– S25.

[61]. Khan KJ, Dubinsky MC, Ford AC, Ullman TA, Talley NJ & Moayyedi P. Efficacy of immunosuppressive therapy for inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol 2011;106:630-642.

ACCEPTED MANUSCRIPT [62]. Su C, Lichtenstein GR, Krok K, Brensinger CM & Lewis JD. A meta-analysis of the placebo rates of remission and response in clinical trials of active Crohn's disease. Gastroenterology. 2004;126:1257-1269.

inflammatory bowel disease. Dig Dis 2009;27 Suppl 1:68-75.

RI PT

[63]. Sands BE. The placebo response rate in irritable bowel syndrome and

[64]. Gallahan WC, Case D & Bloomfeld RS. An analysis of the placebo effect in

SC

Crohn's disease over time. Aliment Pharmacol Ther 2010;31:102-107.

M AN U

*[65]. Fiocchi C. Towards a ‘Cure’ for IBD. Dig Dis 2012;30:428–433.

*[66]. Secher T, Normand S & Chamaillard M. NOD2 prevents emergence of diseasepredisposing microbiota. Gut Microbes 2013;4:353-356.

TE D

[67]. Doherty GA, Bennett GC, Cheifetz AS & Moss AC. Meta-analysis: targeting the intestinal microbiota in prophylaxis for post-operative Crohn's disease. Aliment Pharmacol Ther 2010;31:802-809.

EP

[68]. Cleynen I, Vermeire S. Paradoxical inflammation induced by anti-TNF agents in

AC C

patients with IBD. Nat Rev Gastroenterol Hepatol 2012;9:496-503. [69]. Hampe J, Franke A, Rosenstiel P et al: A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn’s disease in ATG16L1. Nat Genet 2007;39:207-211. [70]. Barnes MJ, Powrie F: Immunology. The gut’s Clostridium cocktail. Science 2011;331:289–290.

ACCEPTED MANUSCRIPT [71]. Duboc H, Rajca S, Rainteau D et al. Connecting dysbiosis, bile-acid dysmetabolism

and

gut

inflammation

in

inflammatory

bowel

diseases.

Gut 2013;62:531-539.

RI PT

[72]. O’Hara AM & Shanahan F. The gut flora as a forgotten organ. EMPO Rep 2006;7:688-693.

[73]. Voutourier-Maillard A, Secher T, Rehman A et al. NOD2-mediated dysbiosis

SC

predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest

M AN U

2013;123:700-711.

[74]. Ha C, Ullman TA, Siegel CA & Kornbluth A. Patients enrolled in randomized controlled trials do not represent the inflammatory bowel disease patient population. Clin Gastroenterol Hepatol 2012;10:1002-1007.

tell

tales

from

TE D

[75]. Kugathasan S, Cohen S: Searching for new clues in inflammatory bowel disease: pediatric

natural

history

studies.

Gastroenterology

EP

2008;135:1038–1041.

IBD

[76]. Reddy SI, Friedman S, Telford JJ et al. Are patients with inflammatory bowel

AC C

disease receiving optimal care? Am J Gastroenterol 2005;100:1357–1361. [77]. Mawdsley JED, Irving PM, Makins RJ, et al. Optimizing quality of outpatient care for patients with inflammatory bowel disease: the importance of specialist clinics. Eur J Gastroenterol Hepatol 2006;18:249–253. *[78]. Crossing the Quality Chasm: A New Health System for the 21st Century. Washington, DC: National Academy Press; 2001.

ACCEPTED MANUSCRIPT

RI PT

Conflicts of interest statement

Dr. Mantzaris has received advisory board fees from Centocor Ortho Biotech, MSD,

SC

Schering-Plough, AbbVie, and Danon; consulting fees from Danon; lecture fees from Ferring International, MSD, AbbVie, FALK Pharma, and Angelini; research support

AC C

EP

TE D

M AN U

from AstraZeneca, AbbVie, Menarini, and Genesis.

When can we cure Crohn's?

Crohn's disease is a life-long idiopathic inflammatory disease which affects the entire gastrointestinal tract and occasionally extra-intestinal organ...
313KB Sizes 1 Downloads 4 Views