Acquired Refractory Iron Deficiency Anemia

Margherita Migone De Amicis1, Alessandro Rimondi2,3, Luca Elli2,3 and Irene Motta1,4

1 General Medicine Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy.
2 Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
3 Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
4 Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.

Correspondence to:  Irene Motta, MD. Department of Clinical Sciences and Community Health, Università degli Studi di Milano,General Medicine Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico. Tel: +390255033493. E-mail: irene.motta@unimi.it

Published: May 1, 2021
Received: October 22, 2020
Accepted: April 6, 2021
Mediterr J Hematol Infect Dis 2021, 13(1): e2021028 DOI 10.4084/MJHID.2021.028

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Anemia is a global health problem affecting one-third of the world population, and half of the cases are due to iron deficiency (ID). Iron deficiency anemia (IDA) is the leading cause of disability in several countries. Although multiple mechanisms may coexist, ID and IDA causes can be classified as i) insufficient iron intake for the body requirement, ii) reduced absorption, and iii) blood losses. Oral iron represents the mainstay of IDA treatment. IDA is defined as "refractory" when the hematologic response after 4 to 6 weeks of treatment with oral iron (an increase of >=1 g/dL of Hb) is absent. The cause of iron-refractory anemia is usually acquired and frequently related to gastrointestinal pathologies, although a rare genetic form called iron-refractory iron deficiency anemia (IRIDA) exists. In some pathological circumstances, either genetic or acquired, hepcidin increases, limiting the absorption in the gut, remobilization, and recycling of iron, thereby reducing iron plasma levels. Indeed, conditions with high hepcidin levels are often under-recognized as iron refractory, leading to inappropriate and unsuccessful treatments. This review provides an overview of the iron refractory anemia underlying conditions, from gastrointestinal pathologies to hepcidin dysregulation and iatrogenic or provoked conditions, and the specific diagnostic and treatment approach.



Introduction

Anemia is a global health problem affecting one-third of the world population, with half of the cases due to iron deficiency (ID).[1] The prevalence of iron deficiency without anemia remains elusive, although it has been estimated that it is at least double that of iron deficiency anemia (IDA). IDA itself is the leading cause of disability in several countries, with children and females of childbearing age being the most affected.[2] Moreover, ID is the cause of anemia in a significant proportion of elderly patients admitted to medical units.[3]
IDA is usually microcytic, defined as a decrease in mean red cell volume (MCV) due to reduced hemoglobin (Hb) production. However, in some circumstances, IDA can be normocytic if folate or vitamin B12 deficiencies coexist.
ID and IDA are the presenting signs and, in some cases, the sole of different medical conditions. ID and IDA causes can be classified as: i) insufficient iron intake for the body requirement, ii) reduced absorption, and iii) blood losses, although multiple mechanisms may coexist. Age, sex, clinical history, and symptoms are essential in guiding the diagnostic work-up for the identification of the underlying cause.[4] Oral iron represents the mainstay of IDA treatment. IDA is defined as "refractory" when the hematologic response after 4 to 6 weeks of treatment with oral iron (an increase of >= 1 g/dL of Hb)[5] is absent. The underlying cause of iron-refractory anemia is usually acquired and frequently related to gastrointestinal (GI) pathologies, although a rare genetic form called iron-refractory iron deficiency anemia (IRIDA) exists (See definitions reported in table 1).


Table 1 Table 1. Definitions related to iron deficiency.

Iron absorption is limited to 1 to 2 mg daily, while most of the iron needed is provided through recycling by macrophages that phagocytize senescent erythrocytes. The two latter mechanisms are controlled by the hormone hepcidin, the master regulator of iron homeostasis.[6] Hepcidin is a peptide primarily produced by the liver,[7] which regulates the systemic flux of iron by modulating ferroportin levels through its degradation.[8] Ferroportin is highly expressed in duodenal enterocytes to transport the iron absorbed with the diet, in hepatocytes to transport stored iron, and macrophages to transport recycled iron. Hepcidin expression is regulated by several mechanisms, including iron status, erythropoietic stimuli, and inflammation. In ID, hepcidin synthesis is suppressed to facilitate the absorption of iron.
However, in some pathological circumstances, either genetic or acquired, hepcidin increases, reducing surface ferroportin, limiting the absorption, remobilization, and recycling of iron, thereby reducing iron plasma levels.[8] Indeed, conditions with high hepcidin levels are often under-recognized as iron refractory, leading to inappropriate and unsuccessful treatments. This review provides an overview of the iron-refractory anemia underlying conditions (Figure 1), from GI pathologies to hepcidin dysregulation and iatrogenic or provoked conditions.


Figure 1 Figure 1. Causes of iron refractory iron deficiency anemia. IRIDA: Iron refractory iron deficiency anemia; IBD: Inflammatory bowel diseases.


 

Autoimmune Atrophic Gastritis

Chronic atrophic gastritis (CAG) is defined as a loss of gastric glands replaced by pseudo-pyloric or intestinal metaplasia or fibrosis, causing impaired gastric acid secretion (hypochlorhydria/achlorhydria) and intrinsic factor deficiency. Historically, two main types of CAG have been documented: type A and type B chronic atrophic gastritis. Type A is associated with autoimmune etiology, anti-parietal cell antibodies (APCA), and corpus involvement with the antrum sparing. On the other hand, type B is usually associated with environmental factors such as Helicobacter Pylori (HP) infection and generally affects antral mucosa.[9]
However, more recent consensus and guidelines have revised the terminology, regrouping CAG by considering morphology, topography, distribution of gastritis, and the possible coexistence of multiple etiologies. Asides from classical dichotomous classification, it is now assumed that histological sampling can hint at unclassified damage to the gastric mucosa, and multiple patterns of gastritis can be found in the same patient. Moreover, an overlap between different types of gastritis (e.g., H. Pylori corpus predominant pattern) has been described in some patients.[10,11] Chronic atrophic gastritis prevalence changes extensively among epidemiological studies as different diagnosis and definition criteria are adopted. However, a systematic review esteems a prevalence of CAG as high as 23.9% in the general population and up to 27% in selected groups when serological criteria, such as pepsinogen I or pepsinogen I/pepsinogen II ratio, are adopted.[12]
Patients with CAG are usually affected by a certain degree of anemia. Vice versa, some studies show that up to 20-27% with refractory IDA are diagnosed with autoimmune gastritis.[13,14] Nonetheless, the burden and clinical effects of CAG are relatively underestimated by GI specialists.[15]
As HP infection is examined separately in this review, henceforth, we will discuss autoimmune chronic atrophic gastritis and its relationship with IDA. According to the literature, roughly 2% of the general population is affected by autoimmune atrophic gastritis (AAG). This illness is usually found in females over 60 years old with related autoimmune diseases.[16] Once established, AAG can be responsible for many pathologic alterations involving multiple apparatuses (e.g., gastrointestinal, hematological, neurological). As AAG affects oxyntic gland production of hydrochloric acid and intrinsic factor, both B12 and iron deficiency anemia are reported in these patients. An acidic environment is essential for the reduction, solubilization, and absorption of iron and intrinsic factor, a well-known element for the assimilation of cobalamin. In this subset of patients, current evidence shows that IDA usually occurs before B12 deficiency. It has also been observed that IDA can be the only sign of AAG at clinical presentation in up to 53% of cases. However, subtle alterations, such as anisocytosis, could hint at an early stage of the disease and should always be considered when evaluating a patient.
Patients with AAG-associated IDA are usually younger (up to 20 years) and female. This occurs probably because the reserves of Vitamin B12 can last for a fair amount of time before macrocytic anemia shows up. Overlapping menstrual losses that worsen iron deficiency could explain why premenopausal women account for a more significant percentage of AAG-associated IDA. On the contrary, older male patients with AAG are often diagnosed with B12 deficiency anemia, probably due to a long-lasting unidentified disease. Some studies report dimorphic anemia as a consequence of both iron and B12 deficiency affecting erythrocyte development.
Several studies showed that AAG could be the only cause for iron-refractory anemia. Thus, when examining a patient with IDA who does not respond well to oral iron supplementation, a thorough interrogation on upper gastrointestinal disturbances is mandatory. These patients are often affected by several unspecific symptoms such as abdominal bloating, pain, and nausea. Sometimes symptoms such as early satiety, postprandial fullness, and epigastric pain are also reported, giving rise to differential diagnosis between other common gastrointestinal diseases (e.g., functional disorders, celiac disease).
Nonetheless, patients without enteric symptoms should also be suspected of gastrointestinal involvement.
Besides, attention should be given to those patients with IDA affected by other autoimmune diseases classically related to AAG (e.g., diabetes mellitus type 1, autoimmune thyroiditis).[13,14,16,17]
To our current knowledge, the mainstays for AAG diagnosis are endoscopic gastric evaluation and mucosal biopsies taken following the Sydney Houston protocol.
This protocol allows a correct gastric evaluation and detects 90% of intestinal metaplasia when attained. APCA and anti-intrinsic factor antibodies (AIFA) were once considered a mainstay of the diagnosis. However, current guidelines report a relatively low sensitivity for AIFA, low specificity for APCA, and an overall low diagnostic accuracy when serological markers are considered alone. The best practice imposes accurate histological examination, whereas the serological test may help in corroborating the diagnosis.[13,17,18] Recent laboratory-based scores taking into account gastrin, hemoglobin, and mean cell volume levels have been proposed for identifying subjects more likely to be affected by AAG. Notably, when proton-pump inhibitor (PPI) use and Zollinger-Ellison syndrome have been excluded, gastrin alone stands out as a good indicator of AAG in the atrophic stage, and markedly elevated levels are associated with type 1 neuroendocrine tumor.[19,20]

Helicobacter Pylori

The relationship between Helicobacter pylori and IDA has always been debated. Chronic gastritis resulting in hypochlorhydria overlapping AAG, occult blood loss due to chronic erosive gastritis and peptic ulcers, reduced ascorbic acid, subsequent intestinal iron absorption, and iron sequestration and utilization by the pathogen itself are the supposed mechanisms for IDA in HP infection.[21] The 2017 Maastricht V/Florence Consensus Report declares that HP association with IDA has been conclusively proven in the adult and pediatric population, thus recommending HP eradication in patients with moderate to severe anemia with negative bidirectional endoscopy.[22] However, the authors stated that the overall level of evidence is very low, and the recommendation grade is weak. Notably, this statement is mainly based on relatively old meta-analyses and multicenter studies. Similarly, the American College of Gastroenterology 2017 Guidelines suggest that patients with unexplained IDA despite an appropriate evaluation should be tested for HP infection. Treatment should be given to those who tested positive. However, there are multiple concerns over HP testing (low positive predicting value in low prevalence scenarios) and extensive treatment.[22,23] Furthermore, thanks to the broader availability of capsule endoscopy, it is no longer generally recommended to stop endoscopic examinations after a negative bidirectional endoscopy and encourage to look for other bleeding sources throughout the small GI.[24]
Moreover, a recent retrospective single cohort study based on many consecutive patients undergoing esophagogastroduodenoscopy and standard testing for HP infection excluded an association with IDA in univariate and multiple logistic regression analyses.[25]
When approaching a patient with IDA at the current state of the art, HP should be regarded as a possible cause, but it should not prevent further examinations. We recommend ruling out more frequent causes of IDA while planning patient follow-up and dismissal.

Celiac Disease

Celiac disease (CeD) is an autoimmune enteropathy triggered by gluten in genetically predisposed subjects carrying HLA DQ2 or DQ8 genes. The duodenum and the proximal small bowel (jejunum) are the gastrointestinal tracts mostly affected by mucosal inflammation.[26] Eventually, mucosal inflammation ends in mucosal atrophy and villa blunting when left untreated, causing nutrients malabsorption.
Anemia and CeD are strictly related since the absorption of these nutrients is necessary for red cell production. Elemental iron and heme compounds are absorbed exactly in the gastrointestinal mucosal area involved in CeD.[17,27]
Several studies and a meta-analysis showed that anemia affects up to 69% of patients diagnosed with CeD, especially adult females. Moreover, up to 4% of patients affected by anemia are diagnosed with CeD if an active serological screening strategy is pursued.[28,29]
Notably, since iron is absorbed in the duodenum, IDA is the most common extra-intestinal sign of CeD. Also, IDA could be the main and only manifestation in relatively asymptomatic celiac patients.[30]
Recent studies showed that there is indeed a relationship between the degree of villous atrophy and iron serological markers. Consequently, severe anemia cases are associated with severe histological patterns of CeD (Marsh-Oberhuber 3b and 3c). Besides, it has been demonstrated that iron regulatory proteins expressed by the enterocyte (e.g., divalent metal transporter 1 DMT1, hephaestin, ferroportin 1 FP1, Transferrin receptor 1 TfR1, and Duodenal cytochrome B Dcytb) are regularly present in CeD, thus denying a possible impaired iron mechanism disorder in this subset of patients.[31,32] Currently, malabsorption is believed to be the primary pathogenic mechanism for IDA in CeD; other genetic factors (e.g., polymorphisms or mutations in TMPRSS6) could worsen the clinical scenario.[33]
For the reasons mentioned above, CeD should always be considered when evaluating a patient with IDA, especially if the patient is young and complains of gastrointestinal symptoms (dyspepsia, bloating, diarrhea, constipation).
Current guidelines suggest this active case-finding policy in patients affected with unexplained IDA or iron refractory IDA.[34-36]
Life-long Gluten-Free Diet (GFD) is the mandatory and most effective treatment for this specific type of IDA. However, in the persistence of IDA or slow repletion of iron storage during GFD, iron supplementation could be needed. This clinical scenario is typically related to higher iron demands in premenopausal women or slow normalization of villous atrophy. In adult patients, this last process could last longer than three years.[37]

Surgical Procedures

Several surgical procedures may lead to ID due to malabsorption. ID is well documented in bariatric surgery, but other surgical procedures can be involved. With the growing prevalence of obesity worldwide, bariatric surgery will be more frequent, given its more significant and sustained improvement in weight loss than non-surgical treatments. Good results on weight also have their side effects, including the risk of nutritional deficiency, with ID as one of the most relevant. According to recent data,[38] postoperatively, iron deficiency prevalence varies between 18 and 53 % after Roux-en-Y gastric bypass and between 1 and 54% after sleeve gastrectomy. The prevalence of ID is also proportional to the follow-up length, and it is higher in premenopausal women.[39,40]
Different factors contribute to iron deficiency development after surgery: the pre-existing predisposition related to obesity itself, since obesity-induced systemic inflammation may increase hepcidin levels and contribute to ID; the iron intake becomes inadequate to major requirement due to a higher blood volume.[41,42]
Several studies proved a reduced iron intake after bariatric surgery, primarily due to increased satiety and reduced appetite with a reduction of micronutrient intake,[43] together with a lower tolerance for red meat38 and poor adherence to dietary indications and recommended supplementations.[44] The digestion and absorption of iron are also affected by the induced alteration in gastrointestinal anatomy and physiology, leading to the reduction of the area involved in iron absorption and reduced hydrochloric acid secretion (essential for the reduction of the ferric ion into its ferrous absorbable form).
Other types of surgery, with various indications, may lead to IDA. These include gastric resection, procedures involving the first tract of the small intestine, and proctocolectomy. A frequent complication in patients undergoing surgery for ulcerative colitis is pouchitis, which is associated with IDA (6-21% of patients with functional pouches) due to mucosal bleeding and impaired iron absorption.[45]
Considering urological surgery, procedures that lead to excluding a bowel segment from the GI may cause metabolic and nutritional disturbances. Folic acid and vitamin B12 deficiency have been described, while the importance of ID is debated: Bambach and Hill described a "frequently found ID after major resection of the small intestine (100±330 cm)",[46] while Salomon et al. did not found any evidence of metabolic disorders, including iron deficiency, after an extended follow-up after Camey type I ileal enterocystoplasty, considering this result possibly related to the short ileal length (35 cm) used in the Camey type I operation.[47] A more recent study, in a population of children and adolescents that underwent ileal urostomy, revealed a reduced level of serum iron and increased total iron-binding capacity (TIBC) in the long term follow up in a small proportion of patients, suggesting as "prudent" to investigate iron parameters in these patients.[48]

Occult Blood Losses

Diaphragmatic hernia and esophagitis. Gastric bleeding from Cameron lesions (linear gastric ulcers or erosions on the mucosal folds at the diaphragmatic impression) is a documented cause of IDA in large diaphragmatic or hiatus hernia;[49,50] however, also axial and para-esophageal hernia, in the absence of Cameron lesions, have been related to IDA.[51] IDA reported incidence for all types of hernia varies from 8% to 42% in different cohorts, with an average of 20%.[52] The presence of hiatal hernia itself, independently of esophagitis, has been shown to increase IDA risk.[53] The hypothesis for these forms of IDA has been related to mechanical trauma plus esophagitis, and erosions, or gastroesophageal acid reflux. Of note, the absence of endoscopically detectable erosions does not exclude their causal role in most patients with hernia-related IDA. Considering the treatment of hernia and esophagitis, surgery, in combination with PPI, did not show better results compared to PPI therapy alone in treating and preventing the recurrence of IDA, even in the case of larger diaphragmatic hernia.[51,52]
Small bowel vascular lesions. Small bowel bleedings account for approximately 5% of all cases of GI bleeding[54] and consist of the majority of obscure gastrointestinal bleeding (OGIB), defined as GI bleeding from an unidentified origin that persists despite a comprehensive upper and lower GI evaluation.[97] Age of onset and detailed medical history, including information on comorbidities, is essential for determining small bowel bleeding origin.
The American College of Gastroenterology guidelines in 2015 included inflammatory bowel disease, Dieulafoy's lesion, neoplasia, Meckel's diverticulum, and polyposis syndromes among the most common causes in subjects under the age of 40, while angioectasia, Dieulafoy's lesion, neoplasia, and non-steroidal anti-inflammatory drugs ulcers in those over the age of 40.[55] Also, rare conditions can cause small bowel bleedings.
Capsule endoscopy (CE) is the first-line diagnostic tool for patients with IDA and suspected obscure midgut bleeding, while device-assisted enteroscopy should be performed as the second-line intervention and in case of operative enteroscopy or bioptic sampling.[30]
Intestinal parasitic infections. Several studies have shown parasitic infections, especially T. trichiura and hookworm infections, to be strongly associated with IDA. Hookworm infections are associated with mucosal damage and endogenous loss of iron, while T. trichiura and E. histolytica cause bleeding and dysentery by invading the mucosa of the large intestine. Given these mechanisms, parasitic infections need to be considered in the presence of IDA, especially if iron -refractory.[56-58]

Drug-Related Iron Refractory Anemia

Hypo- or achlorhydria induced by proton pump inhibitors increase the risk of ID and IDA. A recent population-based case-control study in the United Kingdom showed that continuous use of PPI for one year or longer increased the risk of ID compared to the non-exposed subjects or patients treated for less than one year.[59] Suboptimal response to oral iron treatment in patients with ID receiving a PPI has been documented.[60] Several studies demonstrate that PPIs are overprescribed; thus, more attention is required to evaluate benefit and risk balance.

Self-Induced Bleeding

Lasthénie de Ferjol syndrome (LF) is a rare psychiatric disease, affecting women mainly. It is characterized by severe recurrent IDA caused by repeated episodes of self-induced blood-letting. The microcytic anemia observed in a patient with LF syndrome is non-specific, reflecting only chronic blood loss. The work-up is negative for gastrointestinal or gynecological bleeding and nutritional deficiencies.
Anemia improves with therapy, but patients almost inevitably interrupt the treatment with the reappraisal of blood-letting sessions. Despite severe debilitation, most LF syndrome patients continue to fulfill their professional and social duties; they appear highly cooperative and willingly submit to even the most invasive diagnostic procedures to discover the actual cause of their symptoms. The diagnosis is based on the findings of anemia, together with a unique psychological history. Early diagnosis is usually difficult, but it may prevent repeated hospitalizations and the risks associated with invasive diagnostic procedures; management is usually challenging but should be based on long-term psychotherapy.[61,62]

Iron Refractory Anemia and Hepcidin Dysregulation

High hepcidin levels induce ferroportin degradation, limiting the absorption of iron in the gut and the recycling from the liver and macrophages in the spleen.
Iron-refractory iron deficiency anemia (IRIDA). Iron refractory iron deficiency anemia (IRIDA) is a rare hereditary recessive form of microcytic hypochromic anemia. It is caused by mutations in the transmembrane protease serine 6 (TMPRSS6) gene, encoding for matriptase-2, which lead to inappropriately high hepcidin expression for the low iron status and restricted intestinal iron absorption.[63] Patients present with low transferrin saturation and variable values of serum ferritin. Diagnosis is made through molecular analysis, usually during childhood. The disease is refractory to oral iron treatment but shows a slow response to intravenous iron injections with partial correction of the anemia.[64]
Anemia of chronic disease. Elevated hepcidin levels are among the hallmarks of anemia of inflammation (AI), also known as anemia of chronic disease (ACD). It is estimated that up to 40% of all the anemia cases can be considered AI or with significant AI contribution, accounting for 1 billion affected individuals.[2] Under conditions of chronic inflammation, increased expression of interleukin-6 (IL 6) results in the activation of transcription 3 (STAT3) mediated hepcidin expression and, consequently, limited dietary iron absorption and availability for erythropoiesis.[65,66] AI is usually mild to moderate normochromic and normocytic. Characteristically, AI has reduced circulating iron concentrations and transferrin saturation (TSAT) and reduced reticulocyte count. Serum ferritin can be normal or increased, depending on the underlying condition.[67]
Inflammatory bowel diseases. Anemia is the most common extra-intestinal complication in inflammatory bowel diseases (IBD), with a reported prevalence higher than 70%.[68] The etiology of anemia in IBD is multifactorial, mostly involving ID and chronic inflammation.[68] IDA, the main cause of anemia in these patients, is due to chronic blood loss in the gastrointestinal tract, reduced iron intake, and malabsorption. AI is mediated by inflammatory cytokines, including tumor necrosis factor-α, interleukin-1, IL-6, interleukin-10, and interferon-γ.[69,70] Indeed, hepcidin has been demonstrated to be high in IBD patients.[71-73] Hepcidin levels may vary according to the disease activity and the opposite contribution of inflammatory activity versus iron deficiency.
Chronic kidney disease. Hepcidin is cleared by the kidney, where it is mainly reabsorbed and degraded by the proximal tubular mechanism that metabolizes other peptides. In chronic kidney disease (CKD), iron metabolism is disrupted because of high hepcidin concentrations.[74] Several mechanisms contribute to high hepcidin levels and include the inflammatory pathogenesis of kidney disorder, decreased clearance, and hemodialysis or peritoneal dialysis-related complications.[75,76] In these patients, ID is a consequence of decreased iron absorption and increased iron losses, mostly from gastrointestinal bleeding[77] but also from iatrogenic effects. Indeed, hemodialysis causes blood loss due to residual blood in the dialysis equipment, anticoagulation, and frequent laboratory examinations.[74]
Hepcidin adenomas. A form of microcytic hypochromic iron deficiency anemia, refractory to oral treatment, was described in a cohort of patients affected by type 1a glycogen storage disease (GSD1a), a rare recessive disorder due to deficiency of glucose-6 phosphatase, which catalyzes a reaction involved in both glycogenolysis and gluconeogenesis. This condition is characterized by hypoglycemia, and current treatment leads to prolonged survival of affected subjects up to adulthood, with the development of several complications, including liver adenomas. The observation of IDA correction after adenomas resection led to further studies that showed inappropriately high expression of hepcidin mRNA in adenoma tissue (while hepcidin suppression was documented in the normal liver tissue). The aberrant high-level hepcidin mRNA expression is supposed to play a direct role in anemia pathogenesis, which presents the same characteristics of IRIDA.[78,79]

Diagnostic Approach

Considering the refractory IDA definition, we will focus on specific requirements after the IDA first-line approach has been performed (Figure 2). It can be complicated and even experienced, and trained physicians may be misled by low compliance and factitious anemia. C-reactive protein and other acute-phase markers need to be assessed, together with kidney function; coexisting cobalamin or other vitamin deficiency can help address the diagnosis.

Figure 2 Figure 2. Characteristics, indications, and side effects of oral and IV iron formulation.

Medical and pharmacological history needs to be re-evaluated, focusing on GI symptoms and the use of PPI or other drugs that can impair gastric acidity; occult blood losses can be induced by drugs or by hereditary bleeding disorders. History of travel or other risk factors for parasitic infection has to be considered.
The probability of HP infection is higher in men and postmenopausal women, while refractory IDA stating during childhood can be found in IRIDA patients. A specific approach for each condition is reported in Table 3.

Table 2 Table 2. Characteristics, indications, and side effects of oral and IV iron formulation.

Table 3 Table 3. Diagnostic work-up and treatment of specific conditions causing iron refractory anemia.

Treatment

The basis of IDA treatment is the administration of iron to restore the deposits and induce Hb production, together with the removal of the causes, whenever possible. The mainstay is oral iron administration, which should be prolonged for at least three months to replete iron stores and normalize ferritin
levels. Oral iron is cheap and does not require hospital access. However, it is associated with frequent side effects, especially GI symptoms that often lead to treatment interruption.[80] It has been recently shown that oral alternate-day administration can have fewer gastrointestinal side effects and reduced hepcidin levels than conventional daily dosage, ameliorating iron absorption and improving patient tolerability.[81,82] Thus, in oral treatment failure, low adherence should be suspected, but iron refractoriness causes should also be considered.
Iron can also be administered intravenously (IV), with more rapid and effective correction of ID and IDA bypassing iron absorption. It has higher costs, but with more recent formulation, such as ferric carboximaltose, fewer/single-dose administration is often sufficient, with shorter time for anemia correction and reduced hospital accesses. New IV iron formulations, compared to those available in the past, are safe and exhibit a low immunogenic potential.[83] The cost-benefit ratio favors IV supplementation, as indicated by the European Medicines Agency (EMEA: http://www.emea.europe.eudocument WC500150771, 2013).
Characteristics, indications, and side effects of oral and IV formulation are reported in Table 2. The choice between different formulations is mainly based on Hb values, IDA etiology, comorbidities, and oral formulation tolerance.
The specific treatment approach for each condition is presented in Table 3.

Conclusions

IDA is a highly prevalent disorder affecting all ages. However, it remains under-recognized, and unique definitions of ID, anemia, and IDA are necessary to reach a proper diagnosis.[84] Once diagnosed, IDA requires appropriate diagnostic work-up to identify the underlying cause and start the proper treatment. Oral iron refractory conditions require specialist management and specific treatment to avoid the worsening of IDA and consequences on the patient's quality of life.

References   

  1. Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388(10053):1545-1602. https://doi.org/10.1016/S0140-6736(16)31678-6 
  2. Kassebaum NJ, Jasrasaria R, Naghavi M, Wulf SK, Johns N, Lozano R, Regan M, Weatherall D, Chou DP, Eisele TP, Flaxman SR, Pullan RL, Brooker SJ, Murray CJ. A systematic analysis of global anemia burden from 1990 to 2010. Blood. 2014;123(5):615-624. https://doi.org/10.1182/blood-2013-06-508325 PMid:24297872 PMCid:PMC3907750 
  3. Migone De Amicis M, Poggiali E, Motta I, Minonzio F, Fabio G, Hu C, Cappellini MD. Anemia in elderly hospitalized patients: prevalence and clinical impact. Intern Emerg Med. 2015;10(5):581-586. https://doi.org/10.1007/s11739-015-1197-5 PMid:25633233
  4. Camaschella C. Iron deficiency. Blood. 2019;133(1):30-39. https://doi.org/10.1182/blood-2018-05-815944 PMid:30401704 
  5. Hershko C, Camaschella C. How I treat unexplained refractory iron deficiency anemia. Blood. 2014;123(3):326-333. https://doi.org/10.1182/blood-2013-10-512624 PMid:24215034 
  6. Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B, Kahn A, Vaulont S. Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci U S A. 2001;98(15):8780-8785. https://doi.org/10.1073/pnas.151179498 PMid:11447267 PMCid:PMC37512 
  7. Park CH, Valore EV, Waring AJ, Ganz T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J Biol Chem. 2001;276(11):7806-7810. https://doi.org/10.1074/jbc.M008922200 PMid:11113131 
  8. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, Ganz T, Kaplan J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306(5704):2090-2093. https://doi.org/10.1126/science.1104742 PMid:15514116 
  9. Strickland RG, Mackay IR. A reappraisal of the nature and significance of chronic atrophic gastritis. Am J Dig Dis. 1973;18(5):426-440. https://doi.org/10.1007/BF01071995 PMid:4573514 
  10. Dixon MF, Genta RM, Yardley JH, Correa P. Classification and grading of gastritis. The updated Sydney System. International Workshop on the Histopathology of Gastritis, Houston 1994. Am J Surg Pathol. 1996;20(10):1161-1181. https://doi.org/10.1097/00000478-199610000-00001 PMid:8827022 
  11. Sugano K, Tack J, Kuipers EJ, Graham DY, El-Omar EM, Miura S, Haruma K, Asaka M, Uemura N, Malfertheiner P, faculty members of Kyoto Global Consensus C. Kyoto global consensus report on Helicobacter pylori gastritis. Gut. 2015;64(9):1353-1367. https://doi.org/10.1136/gutjnl-2015-309252 PMid:26187502 PMCid:PMC4552923 
  12. Marques-Silva L, Areia M, Elvas L, Dinis-Ribeiro M. Prevalence of gastric precancerous conditions: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2014;26(4):378-387. https://doi.org/10.1097/MEG.0000000000000065 PMid:24569821 
  13. Lahner E, Zagari RM, Zullo A, Di Sabatino A, Meggio A, Cesaro P, Lenti MV, Annibale B, Corazza GR. Chronic atrophic gastritis: Natural history, diagnosis and therapeutic management. A position paper by the Italian Society of Hospital Gastroenterologists and Digestive Endoscopists [AIGO], the Italian Society of Digestive Endoscopy [SIED], the Italian Society of Gastroenterology [SIGE], and the Italian Society of Internal Medicine [SIMI]. Dig Liver Dis. 2019;51(12):1621-1632. https://doi.org/10.1016/j.dld.2019.09.016 PMid:31635944 
  14. Lenti MV, Lahner E, Bergamaschi G, Miceli E, Conti L, Massironi S, Cococcia S, Zilli A, Caprioli F, Vecchi M, Maiero S, Cannizzaro R, Corazza GR, Annibale B, Di Sabatino A. Cell Blood Count Alterations and Patterns of Anaemia in Autoimmune Atrophic Gastritis at Diagnosis: A Multicentre Study. J Clin Med. 2019;8(11). https://doi.org/10.3390/jcm8111992 PMid:31731715 PMCid:PMC6912578 
  15. Lenti MV, Miceli E, Cococcia S, Klersy C, Staiani M, Guglielmi F, Giuffrida P, Vanoli A, Luinetti O, De Grazia F, Di Stefano M, Corazza GR, Di Sabatino A. Determinants of diagnostic delay in autoimmune atrophic gastritis. Aliment Pharmacol Ther. 2019;50(2):167-175. https://doi.org/10.1111/apt.15317 PMid:31115910 
  16. Zilli A, Cavalcoli F, Ciafardini C, Massironi S. Deficiency of micronutrients in patients affected by chronic atrophic autoimmune gastritis: A single-institution observational study. Dig Liver Dis. 2019;51(4):505-509. https://doi.org/10.1016/j.dld.2018.08.028 PMid:30236765 
  17. Bergamaschi G, Di Sabatino A, Corazza GR. Pathogenesis, diagnosis and treatment of anaemia in immune-mediated gastrointestinal disorders. Br J Haematol. 2018;182(3):319-329. https://doi.org/10.1111/bjh.15254 PMid:29732532 
  18. Banks M, Graham D, Jansen M, Gotoda T, Coda S, di Pietro M, Uedo N, Bhandari P, Pritchard DM, Kuipers EJ, Rodriguez-Justo M, Novelli MR, Ragunath K, Shepherd N, Dinis-Ribeiro M. British Society of Gastroenterology guidelines on the diagnosis and management of patients at risk of gastric adenocarcinoma. Gut. 2019;68(9):1545-1575. https://doi.org/10.1136/gutjnl-2018-318126 PMid:31278206 PMCid:PMC6709778 
  19. Miceli E, Padula D, Lenti MV, Gallia A, Albertini R, Di Stefano M, Klersy C, Corazza GR. A laboratory score in the diagnosis of autoimmune atrophic gastritis: a prospective study. J Clin Gastroenterol. 2015;49(1):e1-5. https://doi.org/10.1097/MCG.0000000000000101 PMid:24583750 
  20. Lenti MV, Rugge M, Lahner E, Miceli E, Toh BH, Genta RM, De Block C, Hershko C, Di Sabatino A. Autoimmune gastritis. Nat Rev Dis Primers. 2020;6(1):56. https://doi.org/10.1038/s41572-020-0187-8 PMid:32647173 
  21. DuBois S, Kearney DJ. Iron-deficiency anemia and Helicobacter pylori infection: a review of the evidence. Am J Gastroenterol. 2005;100(2):453-459. https://doi.org/10.1111/j.1572-0241.2005.30252.x PMid:15667507 
  22. Malfertheiner P, Megraud F, O'Morain CA, Gisbert JP, Kuipers EJ, Axon AT, Bazzoli F, Gasbarrini A, Atherton J, Graham DY, Hunt R, Moayyedi P, Rokkas T, Rugge M, Selgrad M, Suerbaum S, Sugano K, El-Omar EM, European H, Microbiota Study G, Consensus p. Management of Helicobacter pylori infection-the Maastricht V/Florence Consensus Report. Gut. 2017;66(1):6-30. https://doi.org/10.1136/gutjnl-2016-312288 PMid:27707777
  23. Chey WD, Leontiadis GI, Howden CW, Moss SF. ACG Clinical Guideline: Treatment of Helicobacter pylori Infection. Am J Gastroenterol. 2017;112(2):212-239. https://doi.org/10.1038/ajg.2016.563 PMid:28071659
  24. Pennazio M, Spada C, Eliakim R, Keuchel M, May A, Mulder CJ, Rondonotti E, Adler SN, Albert J, Baltes P, Barbaro F, Cellier C, Charton JP, Delvaux M, Despott EJ, Domagk D, Klein A, McAlindon M, Rosa B, Rowse G, Sanders DS, Saurin JC, Sidhu R, Dumonceau JM, Hassan C, Gralnek IM. Small-bowel capsule endoscopy and device-assisted enteroscopy for diagnosis and treatment of small-bowel disorders: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline. Endoscopy. 2015;47(4):352-376. https://doi.org/10.1055/s-0034-1391855 PMid:25826168
  25. John S, Baltodano JD, Mehta N, Mark K, Murthy U. Unexplained iron deficiency anemia: does Helicobacter pylori have a role to play? Gastroenterol Rep (Oxf). 2018;6(3):215-220. https://doi.org/10.1093/gastro/goy001 PMid:30151206 PMCid:PMC6101634
  26. Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol. 1999;11(10):1185-1194. https://doi.org/10.1097/00042737-199910000-00019 PMid:10524652 
  27. Leffler DA, Green PH, Fasano A. Extraintestinal manifestations of coeliac disease. Nat Rev Gastroenterol Hepatol. 2015;12(10):561-571. https://doi.org/10.1038/nrgastro.2015.131 PMid:26260366 
  28. Mahadev S, Laszkowska M, Sundstrom J, Bjorkholm M, Lebwohl B, Green PHR, Ludvigsson JF. Prevalence of Celiac Disease in Patients With Iron Deficiency Anemia-A Systematic Review With Meta-analysis. Gastroenterology. 2018;155(2):374-382 e371. https://doi.org/10.1053/j.gastro.2018.04.016 PMid:29689265 PMCid:PMC7057414 
  29. Corazza GR, Valentini RA, Andreani ML, D'Anchino M, Leva MT, Ginaldi L, De Feudis L, Quaglino D, Gasbarrini G. Subclinical coeliac disease is a frequent cause of iron-deficiency anaemia. Scand J Gastroenterol. 1995;30(2):153-156. https://doi.org/10.3109/00365529509093254 PMid:7732338 
  30. Elli L, Norsa L, Zullo A, Carroccio A, Girelli C, Oliva S, Romano C, Leandro G, Bellini M, Marmo R, Soncini M, Monica F, De Francesco V, Paulon E, Cappellini MD, Motta I, Ferretti F, Orlando S, Mansueto P, Buscarini E, Manfredi G, Agostoni C, Tomba C, Cannizzaro R. Diagnosis of chronic anaemia in gastrointestinal disorders: A guideline by the Italian Association of Hospital Gastroenterologists and Endoscopists (AIGO) and the Italian Society of Paediatric Gastroenterology Hepatology and Nutrition (SIGENP). Dig Liver Dis. 2019;51(4):471-483. https://doi.org/10.1016/j.dld.2019.01.022 PMid:30850345 
  31. Barisani D, Ceroni S, Del Bianco S, Meneveri R, Bardella MT. Hemochromatosis gene mutations and iron metabolism in celiac disease. Haematologica. 2004;89(11):1299-1305. 
  32. Barisani D, Parafioriti A, Bardella MT, Zoller H, Conte D, Armiraglio E, Trovato C, Koch RO, Weiss G. Adaptive changes of duodenal iron transport proteins in celiac disease. Physiol Genomics. 2004;17(3):316-325. https://doi.org/10.1152/physiolgenomics.00211.2003 PMid:15054143 
  33. Elli L, Poggiali E, Tomba C, Andreozzi F, Nava I, Bardella MT, Campostrini N, Girelli D, Conte D, Cappellini MD. Does TMPRSS6 RS855791 polymorphism contribute to iron deficiency in treated celiac disease? Am J Gastroenterol. 2015;110(1):200-202. https://doi.org/10.1038/ajg.2014.354 PMid:25567183 
  34. Elli L, Ferretti F, Orlando S, Vecchi M, Monguzzi E, Roncoroni L, Schuppan D. Management of celiac disease in daily clinical practice. Eur J Intern Med. 2019;61:15-24. https://doi.org/10.1016/j.ejim.2018.11.012 PMid:30528262 
  35. Ludvigsson JF, Bai JC, Biagi F, Card TR, Ciacci C, Ciclitira PJ, Green PH, Hadjivassiliou M, Holdoway A, van Heel DA, Kaukinen K, Leffler DA, Leonard JN, Lundin KE, McGough N, Davidson M, Murray JA, Swift GL, Walker MM, Zingone F, Sanders DS, Group BSGCDGD, British Society of G. Diagnosis and management of adult coeliac disease: guidelines from the British Society of Gastroenterology. Gut. 2014;63(8):1210-1228. https://doi.org/10.1136/gutjnl-2013-306578 PMid:24917550 PMCid:PMC4112432 
  36. Rubio-Tapia A, Hill ID, Kelly CP, Calderwood AH, Murray JA, American College of G. ACG clinical guidelines: diagnosis and management of celiac disease. Am J Gastroenterol. 2013;108(5):656-676; quiz 677. https://doi.org/10.1038/ajg.2013.79 PMid:23609613 PMCid:PMC3706994 
  37. Elli L, Zini E, Tomba C, Bardella MT, Bosari S, Conte D, Runza L, Roncoroni L, Ferrero S. Histological evaluation of duodenal biopsies from coeliac patients: the need for different grading criteria during follow-up. BMC Gastroenterol. 2015;15:133. https://doi.org/10.1186/s12876-015-0361-8 PMid:26467310 PMCid:PMC4604755 
  38. Steenackers N, Van der Schueren B, Mertens A, Lannoo M, Grauwet T, Augustijns P, Matthys C. Iron deficiency after bariatric surgery: what is the real problem? Proc Nutr Soc. 2018;77(4):445-455. https://doi.org/10.1017/S0029665118000149 PMid:29619914
  39. Schijns W, Ligthart MAP, Berends FJ, Janssen IMC, van Laarhoven C, Aarts EO, de Boer H. Changes in Iron Absorption After Roux-en-Y Gastric Bypass. Obes Surg. 2018;28(6):1738-1744. https://doi.org/10.1007/s11695-017-3088-5 PMid:29327182 
  40. Ruz M, Carrasco F, Rojas P, Codoceo J, Inostroza J, Rebolledo A, Basfi-fer K, Csendes A, Papapietro K, Pizarro F, Olivares M, Sian L, Westcott JL, Hambidge KM, Krebs NF. Iron absorption and iron status are reduced after Roux-en-Y gastric bypass. Am J Clin Nutr. 2009;90(3):527-532. https://doi.org/10.3945/ajcn.2009.27699 PMid:19625680 
  41. Ruivard M, Laine F, Ganz T, Olbina G, Westerman M, Nemeth E, Rambeau M, Mazur A, Gerbaud L, Tournilhac V, Abergel A, Philippe P, Deugnier Y, Coudray C. Iron absorption in dysmetabolic iron overload syndrome is decreased and correlates with increased plasma hepcidin. J Hepatol. 2009;50(6):1219-1225. https://doi.org/10.1016/j.jhep.2009.01.029 PMid:19398238 
  42. Aigner E, Feldman A, Datz C. Obesity as an emerging risk factor for iron deficiency. Nutrients. 2014;6(9):3587-3600. https://doi.org/10.3390/nu6093587 PMid:25215659 PMCid:PMC4179177 
  43. Moize V, Andreu A, Flores L, Torres F, Ibarzabal A, Delgado S, Lacy A, Rodriguez L, Vidal J. Long-term dietary intake and nutritional deficiencies following sleeve gastrectomy or Roux-En-Y gastric bypass in a mediterranean population. J Acad Nutr Diet. 2013;113(3):400-410. https://doi.org/10.1016/j.jand.2012.11.013 PMid:23438491 
  44. Hood MM, Corsica J, Bradley L, Wilson R, Chirinos DA, Vivo A. Managing severe obesity: understanding and improving treatment adherence in bariatric surgery. J Behav Med. 2016;39(6):1092-1103. https://doi.org/10.1007/s10865-016-9772-4 PMid:27444752 
  45. M'Koma AE, Wise PE, Schwartz DA, Muldoon RL, Herline AJ. Prevalence and outcome of anemia after restorative proctocolectomy: a clinical literature review. Dis Colon Rectum. 2009;52(4):726-739. https://doi.org/10.1007/DCR.0b013e31819ed571 PMid:19404082 PMCid:PMC4154485 
  46. Bambach CP, Hill GL. Long term nutritional effects of extensive resection of the small intestine. Aust N Z J Surg. 1982;52(5):500-506. https://doi.org/10.1111/j.1445-2197.1982.tb06039.x PMid:6816204 
  47. Salomon L, Lugagne PM, Herve JM, Barre P, Lebret T, Botto H. No evidence of metabolic disorders 10 to 22 years after Camey type I ileal enterocystoplasty. J Urol. 1997;157(6):2104-2106. https://doi.org/10.1016/S0022-5347(01)64685-8 
  48. Abd-el-Gawa G, Abrahamsson K, Norlen L, Hjalmas K, Hanson E. Vitamin B12 and folate after 5-12 years of continent ileal urostomy (Kock reservoir) in children and adolescents. Eur Urol. 2002;41(2):199-205. https://doi.org/10.1016/S0302-2838(01)00032-X 
  49. Cameron AJ, Higgins JA. Linear gastric erosion. A lesion associated with large diaphragmatic hernia and chronic blood loss anemia. Gastroenterology. 1986;91(2):338-342. https://doi.org/10.1016/0016-5085(86)90566-4 
  50. Zullo A, Manta R, De Francesco V, Fiorini G, Lahner E, Vaira D, Annibale B. Cameron lesions: A still overlooked diagnosis. Case report and systematic review of literature. Clin Res Hepatol Gastroenterol. 2018;42(6):604-609. https://doi.org/10.1016/j.clinre.2018.05.002 PMid:29910147 
  51. Carrott PW, Markar SR, Hong J, Kuppusamy MK, Koehler RP, Low DE. Iron-deficiency anemia is a common presenting issue with giant paraesophageal hernia and resolves following repair. J Gastrointest Surg. 2013;17(5):858-862. https://doi.org/10.1007/s11605-013-2184-7 PMid:23515913
  52. Panzuto F, Di Giulio E, Capurso G, Baccini F, D'Ambra G, Delle Fave G, Annibale B. Large hiatal hernia in patients with iron deficiency anaemia: a prospective study on prevalence and treatment. Aliment Pharmacol Ther. 2004;19(6):663-670. https://doi.org/10.1111/j.1365-2036.2004.01894.x PMid:15023168
  53. Crooks CJ, West J, Card TR. Upper gastrointestinal haemorrhage and deprivation: a nationwide cohort study of health inequality in hospital admissions. Gut. 2012;61(4):514-520. https://doi.org/10.1136/gutjnl-2011-300186 PMid:21757448 PMCid:PMC3292712
  54. Longstreth GF. Epidemiology and outcome of patients hospitalized with acute lower gastrointestinal hemorrhage: a population-based study. Am J Gastroenterol. 1997;92(3):419-424.
  55. Gerson LB, Fidler JL, Cave DR, Leighton JA. ACG Clinical Guideline: Diagnosis and Management of Small Bowel Bleeding. Am J Gastroenterol. 2015;110(9):1265-1287; quiz 1288. https://doi.org/10.1038/ajg.2015.246 PMid:26303132
  56. Hesham MS, Edariah AB, Norhayati M. Intestinal parasitic infections and micronutrient deficiency: a review. Med J Malaysia. 2004;59(2):284-293.
  57. Kassebaum NJ, Bertozzi-Villa A, Coggeshall MS, Shackelford KA, Steiner C, Heuton KR, Gonzalez-Medina D, Barber R, Huynh C, Dicker D, Templin T, Wolock TM, Ozgoren AA, Abd-Allah F, Abera SF, Abubakar I, Achoki T, Adelekan A, Ademi Z, Adou AK, Adsuar JC, Agardh EE, Akena D, Alasfoor D, Alemu ZA, Alfonso-Cristancho R, Alhabib S, Ali R, Al Kahbouri MJ, Alla F, Allen PJ, AlMazroa MA, Alsharif U, Alvarez E, Alvis-Guzman N, Amankwaa AA, Amare AT, Amini H, Ammar W, Antonio CA, Anwari P, Arnlov J, Arsenijevic VS, Artaman A, Asad MM, Asghar RJ, Assadi R, Atkins LS, Badawi A, Balakrishnan K, Basu A, Basu S, Beardsley J, Bedi N, Bekele T, Bell ML, Bernabe E, Beyene TJ, Bhutta Z, Bin Abdulhak A, Blore JD, Basara BB, Bose D, Breitborde N, Cardenas R, Castaneda-Orjuela CA, Castro RE, Catala-Lopez F, Cavlin A, Chang JC, Che X, Christophi CA, Chugh SS, Cirillo M, Colquhoun SM, Cooper LT, Cooper C, da Costa Leite I, Dandona L, Dandona R, Davis A, Dayama A, Degenhardt L, De Leo D, del Pozo-Cruz B, Deribe K, Dessalegn M, deVeber GA, Dharmaratne SD, Dilmen U, Ding EL, Dorrington RE, Driscoll TR, Ermakov SP, Esteghamati A, Faraon EJ, Farzadfar F, Felicio MM, Fereshtehnejad SM, de Lima GM, Forouzanfar MH, Franca EB, Gaffikin L, Gambashidze K, Gankpe FG, Garcia AC, Geleijnse JM, Gibney KB, Giroud M, Glaser EL, Goginashvili K, Gona P, Gonzalez-Castell D, Goto A, Gouda HN, Gugnani HC, Gupta R, Gupta R, Hafezi-Nejad N, Hamadeh RR, Hammami M, Hankey GJ, Harb HL, Havmoeller R, Hay SI, Pi IB, Hoek HW, Hosgood HD, Hoy DG, Husseini A, Idrisov BT, Innos K, Inoue M, Jacobsen KH, Jahangir E, Jee SH, Jensen PN, Jha V, Jiang G, Jonas JB, Juel K, Kabagambe EK, Kan H, Karam NE, Karch A, Karema CK, Kaul A, Kawakami N, Kazanjan K, Kazi DS, Kemp AH, Kengne AP, Kereselidze M, Khader YS, Khalifa SE, Khan EA, Khang YH, Knibbs L, Kokubo Y, Kosen S, Defo BK, Kulkarni C, Kulkarni VS, Kumar GA, Kumar K, Kumar RB, Kwan G, Lai T, Lalloo R, Lam H, Lansingh VC, Larsson A, Lee JT, Leigh J, Leinsalu M, Leung R, Li X, Li Y, Li Y, Liang J, Liang X, Lim SS, Lin HH, Lipshultz SE, Liu S, Liu Y, Lloyd BK, London SJ, Lotufo PA, Ma J, Ma S, Machado VM, Mainoo NK, Majdan M, Mapoma CC, Marcenes W, Marzan MB, Mason-Jones AJ, Mehndiratta MM, Mejia-Rodriguez F, Memish ZA, Mendoza W, Miller TR, Mills EJ, Mokdad AH, Mola GL, Monasta L, de la Cruz Monis J, Hernandez JC, Moore AR, Moradi-Lakeh M, Mori R, Mueller UO, Mukaigawara M, Naheed A, Naidoo KS, Nand D, Nangia V, Nash D, Nejjari C, Nelson RG, Neupane SP, Newton CR, Ng M, Nieuwenhuijsen MJ, Nisar MI, Nolte S, Norheim OF, Nyakarahuka L, Oh IH, Ohkubo T, Olusanya BO, Omer SB, Opio JN, Orisakwe OE, Pandian JD, Papachristou C, Park JH, Caicedo AJ, Patten SB, Paul VK, Pavlin BI, Pearce N, Pereira DM, Pesudovs K, Petzold M, Poenaru D, Polanczyk GV, Polinder S, Pope D, Pourmalek F, Qato D, Quistberg DA, Rafay A, Rahimi K, Rahimi-Movaghar V, ur Rahman S, Raju M, Rana SM, Refaat A, Ronfani L, Roy N, Pimienta TG, Sahraian MA, Salomon JA, Sampson U, Santos IS, Sawhney M, Sayinzoga F, Schneider IJ, Schumacher A, Schwebel DC, Seedat S, Sepanlou SG, Servan-Mori EE, Shakh-Nazarova M, Sheikhbahaei S, Shibuya K, Shin HH, Shiue I, Sigfusdottir ID, Silberberg DH, Silva AP, Singh JA, Skirbekk V, Sliwa K, Soshnikov SS, Sposato LA, Sreeramareddy CT, Stroumpoulis K, Sturua L, Sykes BL, Tabb KM, Talongwa RT, Tan F, Teixeira CM, Tenkorang EY, Terkawi AS, Thorne-Lyman AL, Tirschwell DL, Towbin JA, Tran BX, Tsilimbaris M, Uchendu US, Ukwaja KN, Undurraga EA, Uzun SB, Vallely AJ, van Gool CH, Vasankari TJ, Vavilala MS, Venketasubramanian N, Villalpando S, Violante FS, Vlassov VV, Vos T, Waller S, Wang H, Wang L, Wang X, Wang Y, Weichenthal S, Weiderpass E, Weintraub RG, Westerman R, Wilkinson JD, Woldeyohannes SM, Wong JQ, Wordofa MA, Xu G, Yang YC, Yano Y, Yentur GK, Yip P, Yonemoto N, Yoon SJ, Younis MZ, Yu C, Jin KY, El Sayed Zaki M, Zhao Y, Zheng Y, Zhou M, Zhu J, Zou XN, Lopez AD, Naghavi M, Murray CJ, Lozano R. Global, regional, and national levels and causes of maternal mortality during 1990-2013: a systematic analysis for the Global Burden of Disease Study  013. Lancet. 2014;384(9947):980-1004.
  58. Crompton DW, Nesheim MC. Nutritional impact of intestinal helminthiasis during the human life cycle. Annu Rev Nutr. 2002;22:35-59. https://doi.org/10.1146/annurev.nutr.22.120501.134539 PMid:12055337
  59. Tran-Duy A, Connell NJ, Vanmolkot FH, Souverein PC, de Wit NJ, Stehouwer CDA, Hoes AW, de Vries F, de Boer A. Use of proton pump inhibitors and risk of iron deficiency: a population-based case-control study. J Intern Med. 2019;285(2):205-214. https://doi.org/10.1111/joim.12826 PMid:30141278
  60. Ajmera AV, Shastri GS, Gajera MJ, Judge TA. Suboptimal response to ferrous sulfate in iron-deficient patients taking omeprazole. Am J Ther. 2012;19(3):185-189. https://doi.org/10.1097/MJT.0b013e3181f9f6d2 PMid:21150767
  61. Piccillo GA, Miele L, Mondati EG, Scuderi R, Nicolosi M, Forgione A, Gabrieli ML, Cefalo C, Gasbarrini G, Grieco A. Eighteen needles to forget...an unnamed past. J Forensic Leg Med. 2007;14(5):304-306. https://doi.org/10.1016/j.jcfm.2006.07.005 PMid:17055322
  62. Palmer SR, Thanarajasingam G, Wolanskyj AP. 39-year-old woman with an obscure case of anemia. Mayo Clin Proc. 2010;85(1):e1-4. https://doi.org/10.4065/mcp.2008.0721 PMid:20042553 PMCid:PMC2800282
  63. Finberg KE, Heeney MM, Campagna DR, Aydinok Y, Pearson HA, Hartman KR, Mayo MM, Samuel SM, Strouse JJ, Markianos K, Andrews NC, Fleming MD. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA). Nat Genet. 2008;40(5):569-571. https://doi.org/10.1038/ng.130 PMid:18408718 PMCid:PMC3104019
  64. De Falco L, Sanchez M, Silvestri L, Kannengiesser C, Muckenthaler MU, Iolascon A, Gouya L, Camaschella C, Beaumont C. Iron refractory iron deficiency anemia. Haematologica. 2013;98(6):845-853. https://doi.org/10.3324/haematol.2012.075515 PMid:23729726 PMCid:PMC3669438 
  65. Gardenghi S, Renaud TM, Meloni A, Casu C, Crielaard BJ, Bystrom LM, Greenberg-Kushnir N, Sasu BJ, Cooke KS, Rivella S. Distinct roles for hepcidin and interleukin-6 in the recovery from anemia in mice injected with heat-killed Brucella abortus. Blood. 2014;123(8):1137-1145. https://doi.org/10.1182/blood-2013-08-521625 PMid:24357729 PMCid:PMC3931188
  66. Theurl I, Aigner E, Theurl M, Nairz M, Seifert M, Schroll A, Sonnweber T, Eberwein L, Witcher DR, Murphy AT, Wroblewski VJ, Wurz E, Datz C, Weiss G. Regulation of iron homeostasis in anemia of chronic disease and iron deficiency anemia: diagnostic and therapeutic implications. Blood. 2009;113(21):5277-5286. https://doi.org/10.1182/blood-2008-12-195651 PMid:19293425
  67. Weiss G, Ganz T, Goodnough LT. Anemia of inflammation. Blood. 2019;133(1):40-50. https://doi.org/10.1182/blood-2018-06-856500 PMid:30401705 PMCid:PMC6536698
  68. Kulnigg S, Gasche C. Systematic review: managing anaemia in Crohn's disease. Aliment Pharmacol Ther. 2006;24(11-12):1507-1523. https://doi.org/10.1111/j.1365-2036.2006.03146.x PMid:17206940
  69. Desreumaux P, Ernst O, Geboes K, Gambiez L, Berrebi D, Muller-Alouf H, Hafraoui S, Emilie D, Ectors N, Peuchmaur M, Cortot A, Capron M, Auwerx J, Colombel JF. Inflammatory alterations in mesenteric adipose tissue in Crohn's disease. Gastroenterology. 1999;117(1):73-81. https://doi.org/10.1016/S0016-5085(99)70552-4
  70. Strong SA, Pizarro TT, Klein JS, Cominelli F, Fiocchi C. Proinflammatory cytokines differentially modulate their own expression in human intestinal mucosal mesenchymal cells. Gastroenterology. 1998;114(6):1244-1256. https://doi.org/10.1016/S0016-5085(98)70431-7
  71. Oustamanolakis P, Koutroubakis IE, Messaritakis I, Malliaraki N, Sfiridaki A, Kouroumalis EA. Serum hepcidin and prohepcidin concentrations in inflammatory bowel disease. Eur J Gastroenterol Hepatol. 2011;23(3):262-268. https://doi.org/10.1097/MEG.0b013e328343b885 PMid:21285884
  72. Mecklenburg I, Reznik D, Fasler-Kan E, Drewe J, Beglinger C, Hruz P, Swiss IBDCSG. Serum hepcidin concentrations correlate with ferritin in patients with inflammatory bowel disease. J Crohns Colitis. 2014;8(11):1392-1397. https://doi.org/10.1016/j.crohns.2014.04.008 PMid:24825446
  73. Martinelli M, Strisciuglio C, Alessandrella A, Rossi F, Auricchio R, Campostrini N, Girelli D, Nobili B, Staiano A, Perrotta S, Miele E. Serum Hepcidin and Iron Absorption in Paediatric Inflammatory Bowel Disease. J Crohns Colitis. 2016;10(5):566-574. https://doi.org/10.1093/ecco-jcc/jjv242 PMid:26733407 PMCid:PMC4957448 
  74. Babitt JL, Lin HY. Mechanisms of anemia in CKD. J Am Soc Nephrol. 2012;23(10):1631-1634. https://doi.org/10.1681/ASN.2011111078 PMid:22935483 PMCid:PMC3458456
  75. Atkinson MA, Kim JY, Roy CN, Warady BA, White CT, Furth SL. Hepcidin and risk of anemia in CKD: a cross-sectional and longitudinal analysis in the CKiD cohort. Pediatr Nephrol. 2015;30(4):635-643. https://doi.org/10.1007/s00467-014-2991-4 PMid:25380788 PMCid:PMC4336204
  76. Troutt JS, Butterfield AM, Konrad RJ. Hepcidin-25 concentrations are markedly increased in patients with chronic kidney disease and are inversely correlated with estimated glomerular filtration rates. J Clin Lab Anal. 2013;27(6):504-510. https://doi.org/10.1002/jcla.21634 PMid:24218134 PMCid:PMC6807340
  77. Sohal AS, Gangji AS, Crowther MA, Treleaven D. Uremic bleeding: pathophysiology and clinical risk factors. Thromb Res. 2006;118(3):417-422. https://doi.org/10.1016/j.thromres.2005.03.032 PMid:15993929
  78. Weinstein DA, Roy CN, Fleming MD, Loda MF, Wolfsdorf JI, Andrews NC. Inappropriate expression of hepcidin is associated with iron refractory anemia: implications for the anemia of chronic disease. Blood. 2002;100(10):3776-3781. https://doi.org/10.1182/blood-2002-04-1260 PMid:12393428
  79. Pagani A, Nai A, Silvestri L, Camaschella C. Hepcidin and Anemia: A Tight Relationship. Front Physiol. 2019;10:1294. https://doi.org/10.3389/fphys.2019.01294 PMid:31649559 PMCid:PMC6794341
  80. Girelli D, Ugolini S, Busti F, Marchi G, Castagna A. Modern iron replacement therapy: clinical and pathophysiological insights. Int J Hematol. 2018;107(1):16-30. https://doi.org/10.1007/s12185-017-2373-3 PMid:29196967
  81. Moretti D, Goede JS, Zeder C, Jiskra M, Chatzinakou V, Tjalsma H, Melse-Boonstra A, Brittenham G, Swinkels DW, Zimmermann MB. Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women. Blood. 2015;126(17):1981-1989. https://doi.org/10.1182/blood-2015-05-642223 PMid:26289639
  82. Stoffel NU, Cercamondi CI, Brittenham G, Zeder C, Geurts-Moespot AJ, Swinkels DW, Moretti D, Zimmermann MB. Iron absorption from oral iron supplements given on consecutive versus alternate days and as single morning doses versus twice-daily split dosing in iron-depleted women: two open-label, randomised controlled trials. Lancet Haematol. 2017;4(11):e524-e533. https://doi.org/10.1016/S2352-3026(17)30182-5 
  83. Avni T, Bieber A, Grossman A, Green H, Leibovici L, Gafter-Gvili A. The safety of intravenous iron preparations: systematic review and meta-analysis. Mayo Clin Proc. 2015;90(1):12-23. https://doi.org/10.1016/j.mayocp.2014.10.007 PMid:25572192
  84. De Franceschi L, Iolascon A, Taher A, Cappellini MD. Clinical management of iron deficiency anemia in adults: Systemic review on advances in diagnosis and treatment. Eur J Intern Med. 2017;42:16-23. https://doi.org/10.1016/j.ejim.2017.04.018 PMid:28528999
  85. Cappellini MD, Comin-Colet J, de Francisco A, Dignass A, Doehner W, Lam CS, Macdougall IC, Rogler G, Camaschella C, Kadir R, Kassebaum NJ, Spahn DR, Taher AT, Musallam KM, Group IC. Iron deficiency across chronic inflammatory conditions: International expert opinion on definition, diagnosis, and management. Am J Hematol. 2017;92(10):1068-1078. https://doi.org/10.1002/ajh.24820 PMid:28612425 PMCid:PMC5599965 
  86. Jimenez K, Kulnigg-Dabsch S, Gasche C. Management of Iron Deficiency Anemia. Gastroenterol Hepatol (N Y). 2015;11(4):241-250.
  87. Choe YH, Kim SK, Son BK, Lee DH, Hong YC, Pai SH. Randomized placebo-controlled trial of Helicobacter pylori eradication for iron-deficiency anemia in preadolescent children and adolescents. Helicobacter. 1999;4(2):135-139. https://doi.org/10.1046/j.1523-5378.1999.98066.x PMid:10382128
  88. Tolkien Z, Stecher L, Mander AP, Pereira DI, Powell JJ. Ferrous sulfate supplementation causes significant gastrointestinal side-effects in adults: a systematic review and meta-analysis. PLoS One. 2015;10(2):e0117383. https://doi.org/10.1371/journal.pone.0117383 PMid:25700159 PMCid:PMC4336293
  89. Elli L, Ferretti F, Branchi F, Tomba C, Lombardo V, Scricciolo A, Doneda L, Roncoroni L. Sucrosomial Iron Supplementation in Anemic Patients with Celiac Disease Not Tolerating Oral Ferrous Sulfate: A Prospective Study. Nutrients. 2018;10(3). https://doi.org/10.3390/nu10030330 PMid:29522446 PMCid:PMC5872748
  90. Branchi F, Locatelli M, Tomba C, Conte D, Ferretti F, Elli L. Enteroscopy and radiology for the management of celiac disease complications: Time for a pragmatic roadmap. Dig Liver Dis. 2016;48(6):578-586. https://doi.org/10.1016/j.dld.2016.02.015 PMid:27012449
  91. Vallurupalli M, Divakaran S, Parnes A, Levy BD, Loscalzo J. The Element of Surprise. N Engl J Med. 2019;381(14):1365-1371. https://doi.org/10.1056/NEJMcps1811547 PMid:31577880 PMCid:PMC7029838 
  92. Parrott J, Frank L, Rabena R, Craggs-Dino L, Isom KA, Greiman L. American Society for Metabolic and Bariatric Surgery Integrated Health Nutritional Guidelines for the Surgical Weight Loss Patient 2016 Update: Micronutrients. Surg Obes Relat Dis. 2017;13(5):727-741. https://doi.org/10.1016/j.soard.2016.12.018 PMid:28392254
  93. Albaramki J, Hodson EM, Craig JC, Webster AC. Parenteral versus oral iron therapy for adults and children with chronic kidney disease. Cochrane Database Syst Rev. 2012;1:CD007857. https://doi.org/10.1002/14651858.CD007857.pub2 PMid:22258974 
  94. Macdougall IC, White C, Anker SD, Bhandari S, Farrington K, Kalra PA, McMurray JJV, Murray H, Tomson CRV, Wheeler DC, Winearls CG, Ford I, Investigators P, Committees. Intravenous Iron in Patients Undergoing Maintenance Hemodialysis. N Engl J Med. 2019;380(5):447-458. https://doi.org/10.1056/NEJMoa1810742 PMid:30365356
  95. Dignass AU, Gasche C, Bettenworth D, Birgegard G, Danese S, Gisbert JP, Gomollon F, Iqbal T, Katsanos K, Koutroubakis I, Magro F, Savoye G, Stein J, Vavricka S, European Cs, Colitis O. European consensus on the diagnosis and management of iron deficiency and anaemia in inflammatory bowel diseases. J Crohns Colitis. 2015;9(3):211-222. https://doi.org/10.1093/ecco-jcc/jju009 PMid:25518052
  96. Gordon M, Sinopoulou V, Iheozor-Ejiofor Z, Iqbal T, Allen P, Hoque S, Engineer J, Akobeng AK. Interventions for treating iron deficiency anaemia in inflammatory bowel disease. Cochrane Database Syst Rev. 2021;1:CD013529. https://doi.org/10.1002/14651858.CD013529.pub2 PMid:33471939
    ,

[TOP]