Abstract
The autoimmune protocol diet (AIP) is a personalized elimination diet that aims to determine and exclude the foods that might trigger immune responses, leading to inflammation and symptomatology associated with autoimmune diseases. Focusing on gut health and the importance of the gut microbiome in immune regulation and overall well-being, the AIP starts by eliminating foods that might create negative effects on the patients and continues by developing a personalized and tailored diet plan for them. This comprehensive approach aims to mitigate symptoms and improve quality of life of individuals with autoimmune conditions. This review presents and critically appraises current knowledge on the AIP protocol, highlight any oversights, and discuss findings from relevant clinical trials.
Keywords: Inflammatory bowel disease, Hashimoto thyroiditis, Rheumatoid arthritis, Celiac disease, Food allergy, IgG, Food antigen, Diet
Highlights
-
•
The Autoimmune Protocol (AIP) is a diet designed to enhance immune regulation, promote gut health, and maintain hormonal balance.
-
•
Proper implementation of the AIP requires expert guidance, and adverse events remain inadequately understood.
-
•
Research on the AIP is highlighting the need for further clinical studies to support its application.
Abbreviations
- ADHD
Attention Deficit Hyperactivity Disorder
- AIP
Autoimmune Protocol
- anti-TPO
Anti-Thyroid Autoantibodies
- BC
Before Christ
- BMI
Body Mass Index
- DAMP
Damage-Associated Molecular Pattern
- fT3
Free Tri-Iodothyronine
- fT4
Free Thyroxine
- FODMAP
Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols
- HT
Hashimoto Thyroiditis
- IBD
Inflammatory Bowel Disease
- IEC
Intestinal Epithelial Cell
- IgG
Immunoglobulin G
- MSQ
Medical Symptoms Questionnaire
- NSAID
Non-Steroidal Anti-Inflammatory Drug
- PAMPs
Pathogen-Associated Molecular Patterns
- PRRs
Pattern-Recognizing Receptors
- QoL:
Quality of Life
- RA
Rheumatoid Arthritis
- RAPID3
Routine Assessment of Patient Index Data 3
- RMDs
Rheumatic and Musculoskeletal Diseases
- SF-36
36-Item Short Form Health Survey
- TLRs
Toll-Like Receptors
- TSH
Thyroid Stimulating Hormone
1. Introduction
Rheumatic and musculoskeletal diseases range from classical autoimmunity to adaptive immune responses and auto-inflammatory disorders characterized by innate immunity activation and inflammasomes [1]. A complex network of cytokines and receptors is involved in the inflammatory reaction in RMDs, with treatment including therapeutic antibodies targeting disease-specific molecules [2]. Autoimmunity is characterized by self-reactive adaptive immune components, exceeding the threshold of systemic immune protection. The breaking of self-antigen tolerance initiated an attack against the body's cells [3,4]. The burden of autoimmune diseases is on a constant rise [5,6], exhibiting however, a female predominance. The co-manifestation of multiple autoimmune disorders is being attributed to shared pathogenetic mechanisms and genetic predisposition.
Emerging evidence highlights the role of diet in improving quality of life (QoL) and disease-related symptoms [7]. Diet has been established to contribute to the primary and secondary prevention of autoimmune diseases, as nutrients may regulate inflammatory and immune pathways [8]. Attaining an “optimal” or “suboptimal” diet may influence several immune-related physiological responses. For instance, the Western diet has been linked to an increased risk of engaging autoimmunity [9], as it dysregulates the gut barrier, affecting gut homeostasis and immune response [10]. The Autoimmune Protocol Diet (AIP) [11], a strict extension of the Paleolithic (Paleo) diet framework in an elimination manner, gained attention for its postulated ability to mitigate inflammation and improve symptoms of autoimmune diseases.
The AIP is based on the penetration of food antigens due to a dysfunction in the gut barrier, leading to increased permeability (“leaky gut”) [12]. The latter has been implicated in dysmotility and various autoimmune disorders [13]. Paired with microbial dysbiosis and in the presence of genetic susceptibility, it can synergistically act as an environmental trigger for autoimmunity [13]. Bacterial antigens stimulate intestinal immune cells, generating autoreactive cells that enter the systemic circulation and target peripheral organs [14]. At the same time, bacterial antigens can be translocated systematically through lymphatic connections, leading to autoreactive cell formation [14]. However, the classification of microbiota as either “good” or “bad” in a binary manner is misleading, as it does not acknowledge the interaction between the patient's genetic profile and the pre-existing microbiota [15].
2. Literature research
A comprehensive search was performed on MEDLINE/PubMed, clinicaltrials.gov, and the gray literature on June 2024, for human studies on patients with autoimmune diseases, applying the AIP.
3. Principles of the AIP
The AIP provides nutritious, whole foods, important for immune regulation, nutrient homeostasis, gut health, and hormone regulation [11]). It encompasses three phases: elimination, reintroduction, and maintenance (Fig. 1).
Fig. 1.
Phases of the AIP
AIP: Autoimmune Protocol diet.
The gastrointestinal tract contains a series of protective layers that physically separate systemic circulation from external triggers, including mucus, epithelium, and lamina propria [16]. The intestinal epithelium layer consists of a monolayer of intestinal epithelial cells (IECs) connected by tight junction proteins and desmosomes, exercising strict control over permanent molecules [17]. This layer contains T-lymphocytes and innate lymphoid cells, whereas the lamina propria contains myeloid cells and lymphocytes [18,19]. IECs exhibit pattern-recognizing receptors (PRRs) such as toll-like receptors (TLRs) and identify pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) that can originate from food antigens or gut microbiota [20]. Subsequently, proinflammatory mediators, such as cytokines, are activated, leading to differentiation, maturation, and activation of immune system cells [21,22]. Macrophages and dendritic cells engulf molecules and interact with T-cells, leading to their differentiation specifically for this food antigen [23]. In food tolerance, T-cells differentiate into T-regulatory cells, which release anti-inflammatory cytokines and suppress immune responses. In food intolerance, T-cells differentiate into T-effector cells that may trigger a systemic immune response [24].
3.1. Unfolding the AIP
The AIP's rationale is to reduce inflammation, support gut health, and manage autoimmune conditions by eliminating foods that might trigger immune responses and exacerbate symptoms.
3.1.1. Elimination phase
During the first phase, grains, legumes, nightshades, nuts, seeds, dairy, eggs, coffee, and alcohol are completely removed from the diet (Table 1). In addition, all refined sugars, oils, processed foods, food additives, artificial colors, and flavorings are excluded due to their contribution to gut dysfunction. The use of non-steroidal anti-inflammatory drugs (NSAIDs) is also avoided during this phase. Patients are encouraged to consume nutrient-dense whole foods, such as vegetables, fruits, mono- and poly-unsaturated fatty acids, tubers, wild game, poultry, organ, and non-processed meats (Table 1) [11]. Gluten-rich grains are avoided, as their glycoprotein extract, gliadin, is implicated in autoimmunity [25]. This phase spans from 6 weeks to 6 months [11]. This prolonged duration stems from plasma B-cells living from a few days to a few months, while immunoglobin G (IgG) antibodies (mostly associated with immune tolerance [26]) have a half-life of 21 days [27]. B-cells contribute to food allergen tolerance by producing allergen-specific IgG antibodies [11].
Table 1.
AIP food list.
Foods to consume | Foods to avoid |
---|---|
Nose-to-tail grass-fed or wild-caught animal proteins, including meat (beef, bison, chicken, lamb, mutton, pork, turkey, wild game), fowl, organ meats, bone broth, heart, kidney liver, and tongue, fish (anchovies, catfish, cod, halibut, herring, mackerel, mahi, salmon, sardines, snapper, tilapia, trout, tuna), shellfish (clams, crab, crawfish, lobster, mussels, octopus, oysters, prawns, scallops, shrimp, squid) | All grains, eggs (and egg derivatives; mayo), and legumes, such as adzuki beans green beans, black beans, white beans, kidney beans, garbanzo beans, fava beans, peanuts, pinto beans, red beans, lupin, soy, lentils, peas, hummus |
None | Barley, corn, durum, fonio, Job's tears, kamut, millet, oats, rice, rye, sorghum, spelled, teff, triticale, wheat, wild rice, crackers, rice waffles or pasta made with brown rice, teff, millet, quinoa, amaranth, tapioca, buckwheat, and potato flour |
Coconut oil, milk and butter, olive oil, avocado, avocado oil/butter, black/green olives | All nuts and seeds (e.g., almonds, walnuts, cashews, sesame, flax seeds, hazelnuts, hemp seeds, macadamia nuts, pecans, pine nuts, pistachios, chia seeds, etc.), and their derivatives (e.g., canola oil, walnut oil, almond flour/milk, tahini) |
Artichokes | Amaranth, buckwheat, chia, quinoa |
Herbs and spices not derived from seeds, such as cinnamon, turmeric, thyme, oregano, basil, rosemary, turmeric, dill, ginger, sage, chives, basil, bay leaf, peppermint, garlic, cinnamon, thyme, parsley, saffron, mint | Anise, annatto, black caraway, celery seed, coriander, cumin, dill, fennel, fenugreek, mustard, nutmeg |
Fresh fruits and vegetables; apples, apricots, asparagus, bananas, cantaloupes, capers, arrowroot sweet potatoes, greens, broccoli, brussels sprouts, lettuce, cucumbers, zucchini, cabbage, beets, cauliflower, carrots, collard greens, cassava (tapioca, kale yuca), celery, cherries, cucumbers, dates, figs, honeydew melons kohlrabi, jicama, mustard greens, pumpkins, cabbage, squash, radishes, rutabagas, kiwis, mangoes, nectarines, papayas, peaches, pears, pineapples, plantains, plums pomegranates, watermelons, zucchini, radicchio, turnips, taro, watercress, yams, berries, melon, okra, clementines, grapefruit, lemons, limes, mandarin oranges, oranges, chives, garlic, leeks, onions scallions shallots spring onions etc. | Ashwagandha, bell peppers (sweet peppers), cayenne peppers, cape gooseberries, eggplant, garden, goji berries, hot peppers (chili peppers), naranjillas, paprika, pepinos, pimentos, potatoes tamarillos, tomatillos, tomatoes, ketchup |
Non-dairy fermented foods, including water-kefir, sauerkraut, kombucha, pickled veggies, apple cider vinegar, red wine vinegar, and kimchi | All dairy products, including yogurt, ghee butter, kefir, milk, cheese, and cream |
Honey, maple syrup, date sugar, coconut sugar, molasses, dried fruits | Granulated sugar, added or artificial sweeteners (acesulfame potassium, aspartame, erythritol, mannitol, neotame, saccharin, sorbitol, stevia, sucralose, xylitol), sugar-sweetened beverages, sugary soda drinks, food additives, emulsifiers, food chemicals, agave, barley malt, barley malt syrup, brown rice syrup, brown sugar, cane crystals, cane sugar (refined), caramel, corn syrup, crystalline fructose, fructose, fruit juice, fruit juice concentrate, galactose, glucose, glucose solids, golden syrup, high- fructose corn syrup, invert sugar, inulin, lactose, malt syrup, maltodextrin, maltose |
Herbal teas, green tea, chamomile | Alcohol, beer, wine, and coffee |
AIP: Autoimmune protocol diet.
3.1.2. Reintroduction phase
During the second phase, eliminated foods are reintroduced to identify the ones that trigger individual responses. Generally, there is no rule of thumb on how to initiate the reintroduction. The most common manner is to reintroduce the foods that each patient enjoys the most, or the ones that are less likely to induce negative responses, in an effort to increase the food options. With this in mind, foods have been categorized into four groups based on their likelihood of being well-tolerated [11]. Group 1 consists of egg yolks, legumes, seed oils, and nut oils [11]. Group 2 includes nuts and seeds, cocoa, egg whites, and alcohol [11]. Group 3 comprises cashews and pistachios, eggplant, coffee, and fermented dairy, while Group 4 includes all dairy, white rice, nightshades, alcohol, and gluten-free grains [11]. This phase is time-consuming; if performed methodically however, it results in distinct beneficial health outcomes for each individual.
3.1.3. Maintenance phase
The last AIP phase involves maintaining the protocol and has no specific duration. It aims to provide a healthy diet and lifestyle that will reduce autoimmune responses. In this manner, each patient adopts a dietary pattern associated with a lack of intolerances, considering the reintroduction phase responses.
3.2. Going beyond the paleo diet
The Paleo diet originated in the Paleolithic age, at the time of the primitive Homo sapiens, who relied mainly on hunting for food [28]. Thus, the Paleo diet was based on grass-fed, pasture-raised and game meat, small fish, eggs, fruit, leafy vegetables, nuts, and seeds, according to the geographic region [29]. It consists of a whole-food diet, with nutritionally dense foods of high quality. The Paleo diet is potentially beneficial to metabolic syndrome [30], cardiovascular diseases [31], cancer [32], insulin resistance [33], type 2 diabetes [34], and body composition [35]. Several foods are eliminated during its implementation, including processed foods, added sugars (except for honey), artificial sweeteners, grains, dairy, and legumes [36]. Thus, the Paleo diet consists of a specific, horizontal dietary pattern, similar for each patient. On the contrary, the AIP is personalized, with different foods allowed or eliminated, depending on individual tolerance. Both diet patterns include an elimination phase, but the AIP goes a step beyond the Paleo diet, aiming to minimize the existence of any possible antigen that might trigger an autoimmune response [11,37]. Thus, individuals who follow the AIP will develop a personalized dietary pattern based on their food tolerances. Moreover, accurately implementation of the AIP requires guidance and support, as it is crucial to correctly follow the phases to address individual food sensitivities and health concerns. In contrast, the Paleo diet does not require this level of expert guidance and assistance. Furthermore, the AIP elimination phase is stricter, with more foods being excluded than the Paleo diet, including eggs, nuts and seeds, nightshades, food additives, refined oils, coffee, and alcohol [11].
3.3. The elimination timeline
The basis for elimination diets dates back (Fig. 2) to the Paleolithic era due to the need to search for food and the limited options available at different times [28]. The subsequent reference to food elimination was made by Hippocrates in 400 BC, referring to lactose intolerance, suggesting eliminating dairy products to relieve discomfort [38]. The term “elimination diets” was first reported in 1926 by Dr. Rowe in 1944 [39]. During the same period, the elimination of gluten among patients with celiac disease was reported [40]. In the early 2000s, the fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet emerged as another iteration of elimination diets recommended for alleviating symptoms of irritable bowel syndrome [41]. Over the past decade, specific forms of elimination diets, including the AIP and the Wahls protocol [42], appeared and were recommended for autoimmune diseases and multiple sclerosis, respectively. There is a concerted effort and need to perform clinical studies to ascertain AIP's clinical efficacy accurately.
Fig. 2.
Timeline of the AIP development
AIP: Autoimmune Protocol Diet, B.C.: Before Christ, FODMAPs: Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols.
4. Clinical insights
4.1. Evidence on AIP applications
Elimination diets have long been used to manage diseases, including celiac disease, allergies, and inflammatory bowel disease (IBD). Regarding autoimmune diseases, the AIP diet has been implemented in organ-specific and systemic autoimmune diseases such as Hashimoto thyroiditis (HT), IBD and rheumatoid arthritis (RA), improving QoL and disease-related symptoms [[43], [44], [45]]. Primary studies implementing the AIP in autoimmune diseases are presented in Table 2.
Table 2.
Primary studies implementing the AIP.
First author | Origin | Design | Diagnosis | Participants | Intervention(s) | Intervention duration | Outcomes | Results |
---|---|---|---|---|---|---|---|---|
Abbott [44] | USA | UCT | HT | N = 16, women with overweight/normal BW and HT, age 20–45 years | Online health coaching on a phased elimination diet (AIP) | 10 wk | QoL (SF-36) and clinical symptom burden (MSQ), anthropometry (BW, BMI), thyroid function (TSH, fT3, fT4, TGA, TPO), inflammation (hs-CRP), CBC | Improvement in QoL, regarding the physical role functioning, emotional role functioning, vitality, mental health, social role functioning, bodily pain and general health subscales. The clinical symptom burden decreased post-intervention. No changes in thyroid function. Inflammation (hs-CRP) decreased. BMI and BW decreased. |
Ihnatowicz [43] | Poland | UCT | HT | N = 28, adults with HT (including 1 male) | AIP | 12 wk | Dietary intake, anthropometry, HT symptoms, thyroid function (TSH, fT3, fT4, TPO, TGA, thyroid US) | Following the AIP, the number of people reporting symptoms of malaise decreased, fT3 and fT4 concentrations decreased, remaining within reference range. Slight reduction in TSH and TGA; TPO showed an upward trend. Decrease in thyroid gland volume. Decrease in BW and muscle mass. |
Konijeti [[45], [46], [47]] | USA | UCT (NCT03-512,327) | IBD (UC and CD) | N = 15, patients with IBD, erosions on endoscopy and/or elevated FC, mean disease duration 19 years, active biologic use in 47 % | AIP | 6 wk AIP, followed by a 5-wk maintenance phase | CRP, partial Mayo score, HBI, FC, mucosal RNA expression (RNA-seq on colonic tissue biopsy), endoscopic picture (SES-CD, Rutgeerts and Mayo endoscopy subscore) | Nutrient repletion was initiated for deficiencies in vitamin D (n = 3) and Fe (n = 6). Clinical remission in 73 % by wk 6, maintained during maintenance phase. In UC, partial Mayo score improved from 5.8 to 1.2 (wk 6) and 1.0 (wk 11). In CD, HBI improved from 6.7 to 3.3 (wk 6) and 3.4 (wk 11). CRP and FC did not change. In patients with follow-up endoscopy (wk 11), improvements in Mayo endoscopic (n = 4), SES-CD (n = 1), and Rutgeerts score (n = 1). Modulation of pathways involved in inflammation, metabolic processes, DNA repair, and cellular proliferation, indicating cellular responses to the AIP. |
McNeill [48,49] | New Zealand | Cross-over, non-randomized | RA | N = 9, adults with RA | (i) AIP (ii) control: usual diet |
12 wk AIP (i) 4 wk control (ii), 8 wk AIP (i) |
QoL, RAPID3, RAID, fatigue, pain, sleep, emotional wellbeing | RAPID3 reduced from baseline to wk 12. On AIP, 4 patients reached a clinically meaningful reduction in RAPID3 of >1.2 and endpoint <2. Fatigue, sleep, and pain improved at wk 12. |
AIP: Autoimmune protocol diet; BW: body weight; BMI; Body mass index; CBC: complete blood count; CD: Crohn's disease; CRP: C-reactive protein; DNA: deoxyribonucleic acid; FC: fecal calprotectin; fT3: free T3; fT4: free T4; HBI: Harvey Bradshaw Index; hs-CRP: high-sensitivity C-reactive protein; HT: Hashimoto's thyroiditis; IBD: inflammatory bowel disease; MSQ: Cleveland Clinic Center for Functional Medicine's Medical Symptoms Questionnaire; QoL: quality of life; RA: rheumatoid arthritis; RAID: Rheumatoid arthritis Impact of Disease; RNA: Ribonucleic acid; RAPID3: Routine Assessment of Patient Index Data 3; SES-CD: simple endoscopic score for Crohn's disease; SF-36: 36-Item Short Form Health Survey; TGA: anti-thyroglobulin antibodies; TPO: thyroid peroxidase antibodies; TSH: thyroid stimulating hormone; UC: ulcerative colitis; UCT: uncontrolled clinical trial; US: ultrasound, USA: United States of America, wk: week.
In HT, two uncontrolled clinical trials [43,44] concluded that patients undergoing the AIP improved and showed that the physical and emotional statuses had less malaise and enhanced general health subscales. In the first study, Field [47] studied women with HT who underwent a 10-week online AIP protocol; in the second [43], they implemented the AIP diet for 12 weeks. Both studies reported improvement in physical and emotional well-being, harnessing patient-reported outcome measures such as the 36-Item Short Form Health Survey (SF-36) and the Cleveland Clinic Center for Functional Medicine's Medical Symptoms Questionnaire (MSQ), as well as open-ended questions regarding malaise levels and disease-related symptoms. Objective findings, such as thyroid function, were assessed in both studies; the second [44] reported no changes, whereas the first reported decreased thyroid gland volume and lower free tri-iodothyronine (fT3) and free thyroxine (fT4) concentrations. At the same time, anti-thyroid autoantibodies (anti-TPO) increased, and thyroid stimulating hormone (TSH) concentrations decreased. Since fT3 and fT4 concentrations were reduced, while TSH and anti-TPO concentrations remained unchanged, adherence to the AIP may worsen thyroid function. Inflammatory markers improved significantly post-intervention [44]. Regarding the anthropometric indices, the first study [43] reported a decrease in body mass index (BMI), body fat (as a % of body weight) and muscle mass, indicating a calorie deficit. In contrast, the second [44] recorded a decrease in body weight and BMI. The same study [44] complemented the AIP with lifestyle modifications such as promoting support systems, sleep hygiene, stress management, movement, and increased time spent outdoors, which could have impacted the results.
In IBD, one uncontrolled trial [[45], [46], [47]] implemented the AIP in patients with active disease for 11 weeks. Clinical remission and endoscopic improvement were documented in most patients as opposed to inflammatory markers (C-reactive protein, fecal calprotectin), which remained unaltered. Based on further analysis, treatment with biological therapy did not affect outcomes. On the other hand, vitamin D and iron supplementation may have influenced the results.
In systemic autoimmune diseases, a non-randomized, crossover trial was conducted in patients with RA, comparing QoL measures following the AIP against the usual diet [48,49]. Preliminary results revealed improved sleep, fatigue, pain sensation, and improved patient-reported outcomes related to disease severity (Routine Assessment of Patient Index Data 3–RAPID3).
Table 3 illustrates the outcomes of the primary studies mentioned above. Adverse events reported in the studies above are listed in Table 4. The HT studies [43,44] had no moderate-to-severe adverse events, although some participants had mental challenges during the elimination phase. In the IBD trial [[45], [46], [47]], one participant suffered from partial small-bowel obstruction requiring hospitalization and another mentioned worsening symptoms and withdrew from the study.
Table 3.
Reported adverse events associated with adherence to the AIP.
First author | AEs assessed | AEs reported |
---|---|---|
Abbott [44] | Nutrient deficiencies | Deficiencies in folate, vitamin B12, and B2 |
Ihnatowicz [43] | Any AE | None reported |
Konijeti [[45], [46], [47]] | Partial small bowel obstruction, exacerbation of symptoms associated with an ileocecal valve stricture | 1/9 patient with CD with an ileocecal valve stricture experienced symptom aggravation and was unable to finish the AIP 1/9 patients with CD with postoperative recurrence of ileal involvement experienced partial small bowel obstruction attributable to excessive intake of raw vegetables |
McNeill [48,49] | Any AE | None reported |
AE: adverse event; AIP: Autoimmune protocol diet; CD: Crohn's disease.
Table 4.
Outcomes reported in studies evaluating the effects of the AIP in patients with autoimmune diseases.
First Author | Outcome category | Outcomes | Pre-AIP | Post-AIP | p-value |
---|---|---|---|---|---|
Abbott [44] |
PRO (SF-36 subscales with score 0–100)f | Physical Functioning | 80 (29)i | 95 (10)i | 0.0001 |
Physical Role Functioning | 25 (88)i | 100 (50)i | 0.001 | ||
Emotional Role Functioning | 33 (92)i | 78 (19)i | 0.0063 | ||
Vitality | 23 (19)i | 58 (34)i | <0.0001 | ||
Mental Health | 54 (25)i | 78 (19)i | <0.0001 | ||
Social Role Functioning | 63 (22)i | 81 (22)i | 0.0057 | ||
Bodily Pain | 68 (22)i | 78 (21)i | 0.0112 | ||
General Health | 40 (26)i | 70 (35)i | <0.0001 | ||
PRO (0–100) | MSQb | 92 ± 25h | 29 ± 20h | <0.0001 | |
Inflammation | hs-CRP (mg/dL) | 1.6 ± 1.7h | 1.2 ± 1.3h | 0.0219 | |
WBC (103/μL) | 5.6 ± 1.4h | 5.1 ± 1.4h | NS | ||
Thyroid function | TSH (μIU/mL) | 2.0 ± 1.5h | 2.0 ± 1.4h | NS | |
fT3 (pg/mL) | 2.4 ± 0.6h | 2.4 ± 0.5h | NS | ||
fT4 (ng/dL) | 1.3 ± 0.4h | 1.4 ± 0.4h | NS | ||
TPO (IU/mL) | 225 ± 178h | 219 ± 186h | NS | ||
TGA (IU/mL) | 110 ± 261h | 124 ± 293h | 0.176 | ||
Anthropometric |
Body weight (lbs) | 149.5 ± 19.5h | 143.4 ± 16.7h | 0.002 | |
BMI (kg/m2) |
24.9 ± 2.6h |
23.9 ± 2.2h |
0.002 |
||
Ihnatowicz [43] |
Thyroid function | TSH (mIU/L) | 3.7 ± 4.1h | 2.7 ± 21.6h | NS |
fT3 (pmol/L) | 3.3 ± 0.4h | 2.9 ± 0.8h | 0.009 | ||
fT4 (ng/dL) | 1.4 ± 0.3h | 1.2 ± 0.3h | 0.044 | ||
Anti-TPO (IU/mL) | 210 ± 204h | 294 ± 301h | NS | ||
Anti-TG (IU/mL) | 317 ± 272h | 301 ± 220h | NS | ||
Thyroid US | pp-usg (mL) | 7.2 ± 3.9h | 6.8 ± 3.2h | 0.020 | |
Pt-usg (mL) | 5.3 ± 2.7h | 5.0 ± 2.3h | 0.014 | ||
Anthropometric |
Body weight (kg) | 69.0 ± 12.1h | 65.5 ± 10.8h | <0.0001 | |
BMI (kg/m2) | 25.6 ± 369h | 24.3 ± 3.1h | <0.0001 | ||
Body fat (% weight) | 33.0 ± 7.6h | 29.4 ± 8.0h | <0.0001 | ||
Body muscle (% weight) |
23.0 ± 8.0h |
21.0 ± 5.0h |
0.032 |
||
Konijeti [[45], [46], [47]] |
Disease activity (CD) | HBI (1–25)a | 6.7 ± 1.5h | 3.4 ± 2.6h | 0.004 |
Abdominal pain (0–3) | 0.6 ± 0.5h | 0.6 ± 0.8h | NS | ||
Bowel movement (n of liquid/soft stools) | 3.4 ± 2.2h | 2.4 ± 1.3h | NS | ||
General well-being (0–4) | 1.6 ± 0.5h | 0.3 ± 0.8h | 0.022 | ||
Disease activity (UC) | Partial Mayo score (0–9)c | 5.8 ± 1.2h | 1.0 ± 2.0h | 0.007 | |
Stool frequency (0–4) | 2.0 ± 0.9h | 0.2 ± 0.4h | 0.012 | ||
Rectal Bleeding (score 0–4) | 1.8 ± 0.8h | 0.3 ± 0.8h | 0.017 | ||
Quality of life (UC) | PGA (score 0–10)d | 2.0 ± 0h | 0.5 ± 0.8h | 0.007 | |
SIBDQ (score 10–70)g | 46.5 ± 12.5h | 60.5 ± 4.8h | 0.045 | ||
Inflammation |
Fecal calprotectin (μg/g) | 471 ± 562h | 112 ± 104h | NS | |
CRP (mg/L) |
3.9 ± 5.2h |
3.4 ± 5.3h |
NS |
||
McNeill [48,49] | PRO | RAPID3 (0–30)e | 2.5 [0.7–5.3]j | 1.0 [0–2.5]j | NR |
Fatigue (VAS 0–10) | 4.4 [0–10]j | 1.1 [0–4]j | NR | ||
Sleep (VAS 0–10) | 3.8 [0–10]j | 1.2 [0–2]j | NR | ||
Pain (VAS 0–10) | 3.3 [0–4.5]j | 1.4 [0–3.5]j | NR |
μL: microliter; μΙU: micro-international units; BMI; Body mass index; CBC: complete blood count; CD: Crohn's disease; CRP: C-reactive protein; dL: deciliter; fT3: free T3; fT4: freeT4; g: gram; HBI: Harvey Bradshaw Index; hs-CRP: high-sensitivity C-reactive protein; IU: international units; kg: kilogram; L: Liter; lbs: libra; mg: milligram; mIU: milli-international units; mL: milliliter; MSQ: Cleveland Clinic Center for Functional Medicine's Medical Symptoms Questionnaire; ng: nanogram; NR: not reported; NS: not significant; pg: picogram; PGA: Physician global assessment; pmol: picomole; PRO: Patient-reported outcome; RAPID3: Routine Assessment of Patient Index Data 3; SF-36: 36-Item Short Form Health Survey; TGA: anti-thyroglobulin antibodies; TPO: thyroid peroxidase antibodies; TSH: thyroid stimulating hormone; UC: ulcerative colitis; US: ultrasound; VAS: visual analogue scale; WBC: White blood cells; wk: week.
HBI scores from 1 to 25: <5 remissions; 5–7 mild activity; 8–16 moderate activity; >16 severe activity and includes abdominal pain, bowel movement, and general well-being.
MSQ scores from 0 to 100: Optimal score: less than 10; Mild toxicity: 10–50; Moderate toxicity: 50–100; Severe toxicity: over 100.
Partial Mayo Score scores from 0 to 9: <2 remission; 2–4 mild activity; 5–7 moderate activity; >7 severe activity and includes stool frequency and rectal bleeding.
PGA scores from 0, defined as the absence of disease activity, to 10, defined as maximum disease activity.
RAPID-3 scores from 0 to 30: scores >12 high disease activity; > 6–12 moderate disease activity; > 3–6 low disease activity, and ≤3 remission.
SF-36 scores are between 0 and 100 with 100 representing the highest level of functioning possible.
SIBDQ scores from 0 to 70; 10–45 severely impaired QoL; 45–60 moderately impaired QoL; 60–70 slightly impaired QoL.
Mean ± SD.
Median (IQR).
Mean [range].
4.2. Nutrient supplementation in AIP
Despite its stringent nature, the AIP advocates a nutritious regimen, rich in whole foods, including colorful vegetables and fruits, organic meat, and lean fish (Table 1). This approach ensures that individuals consume more nutrient-dense foods, possibly leading to greater vitamin and nutrient intakes than when on a standard Western diet. Despite the absence of extensive clinical studies examining possible nutrient deficiencies, the data focusing on specific vitamin deficiencies [44,46] are contentious. While the AIP includes some sources of meat and vegetables, excluding grains might result in deficiencies in vitamins B1, B2 and B3, fiber and iron [50]. The study on women with HT [44] revealed that 50 % of the participants exhibited folate, vitamin B12, or riboflavin deficiencies. Moreover, the Paleo diet may hinder adequate vitamin D and calcium intake [51], given the exclusion of vitamin D and calcium-rich dairy products [52,53]. Of course, the degree of nutrient deficiencies greatly depends on how many foods are excluded from an individual's diet, with more strict patterns offering less variety in the nutrient intake and, thus, a greater chance for nutrient deficiencies. Therefore, it would be false to state that all patients following the AIP have the same chances of developing nutrient deficiencies. Nonetheless, there is a need for comprehensive and well-designed studies to ascertain whether the implementation of the AΙP results in deficiencies, or if it necessitates adjuvant oral nutrient supplementation.
4.3. AIP limitations
The AIP research has limitations, such as adverse events (Table 4). In the study among patients with IBD [[45], [46], [47]], one patient with an ileocecal valve stricture experienced symptom aggravation and failed to complete the protocol. In the same protocol, a patient with postoperative recurrence of ileal Crohn's Disease required hospitalization due to partial small bowel obstruction that was caused by high consumption of raw vegetables, salads, and meat and lack of guidance by a registered dietitian [[45], [46], [47]]. Both patients had ileal strictures, sparking discussion regarding implementing AIP in patients with anatomic variations. As mentioned, nutrient deficiencies consist of a limitation when adhering to the AIP, as deficiencies in folate, vitamin B12, or riboflavin were apparent among women with HT [44], emphasizing the need for individualization and the implementation of a well-designed AIP plan delivered by a registered dietitian [44]. Notably, while some deficiencies were present at the study's onset, others were resolved by its conclusion. In addition, to implement the protocol correctly, the patient needs to be in full remission, as the basis of this diet regime is to detect immune responses based on the symptomatology [11]. Thus, if the reintroduction phase occurs while the patients are on a flare, they might be unable to determine the exact foods causing them dysfunction and, finally, observe a beneficial effect.
There is a lack of evidence to suggest the implementation of the AIP in populations with greater nutritional needs, such as pregnant and lactating women and children. In parallel, people with eating disorders or those who are prone to orthorexia may find the nature of the AIP restrictive and potentially triggering for disordered eating behaviors. Those with certain medical conditions and very specific guidelines regarding food intake are also warranted against following the AIP. People with allergies to foods allowed in the general AIP should aim for the additional exclusion of those prompting allergic reactions (IgG exclusion foods). Likewise, patients with malabsorption syndromes or ileal strictures should not follow the AIP due to its high fiber content.
Notably, some primary studies presented herein excluded patients with common comorbidities such as hypertension and did not allow for the intake of other medications, not allowing for external generalization (Table 5). An important limitation is that all studies are grossly underpowered, using subjective rather than objective measures. More studies are required to assess the effect of the AIP on the pharmacokinetics and pharmacodynamics of commonly used medications.
Table 5.
Inclusion and exclusion criteria of the studies investigating the effects of AIP in patients with autoimmune diseases.
First author | Inclusion criteria | Exclusion criteria |
---|---|---|
Abbott [44] | HT diagnosis, English-speaking women, age 20–45 years, BMI 18.4–29.9 kg/m2 | No definite diagnosis of HT, pregnancy, breastfeeding 6 months postpartum, hypertension, diabetes, heart disease, heart failure, liver failure, chronic renal disease, use of other medications (except FDA approved thyroid replacement medications), outside of pre-defined age and BMI limits |
Ihnatowicz [43] | HT diagnosis, age 19–50 years, BMI >19 kg/m2 | Pregnancy, breastfeeding, celiac disease, renal failure, liver failure, advanced atherosclerotic disease, malnutrition, eating disorders, use of an elimination diet the previous year, implanted electro-stimulators |
Konijeti [[45], [46], [47]] | Symptomatic CD (defined as Harvey–Bradshaw index >5) or UC (partial Mayo clinic score >3), objective evidence of active disease (endoscopy or elevated fecal calprotectin), Facebook account, comfortable with e-mails | Pregnancy, breastfeeding, celiac disease, history of positive TTG antibodies, untreated infection, stoma or J-pouch, bowel surgery 12 wks before enrollment, tube or enteral feeding, elemental diet or parenteral alimentation 4 wks before initiation |
McNeill [48,49] | Stable medication and supplements for >8 wks, unrestricted diet | NR |
AIP: autoimmune protocol diet; BMI: Body mass index; CD: Crohn's disease; FDA: Food and Drug Administration; HT: Hashimoto's Thyroiditis; NR: not reported; TTG: anti-tissue transglutaminase; UC: ulcerative colitis; wks: weeks.
Apart from the concerns raised above, additional constraints exist regarding the AIP's implementation. People following the diet require comprehensive guidance to adopt a nutritionally balanced diet that avoids deficiencies. Furthermore, they will need assistance determining the duration of the elimination phase, beginning the reintroduction phase, sequencing the introduction of new foods, and how many days they should wait for the rechallenge. Many individuals report withdrawal symptoms like headaches, fatigue, irritability, or skin flare-ups, which may last for several days to a week, and they generally tend to feel worse before they feel better, as they might eliminate foods like caffeine, that might cause headaches or migraine in its absence. Additionally, the restrictive nature of the AIP may lead to feelings of social isolation and limitation, impacting individuals’ social lives. The AIP is not a dietary pattern that can be implemented for life; this is not its rationale. Remaining in the elimination phase can put an individual at risk for nutrient deficiencies and potential malnutrition. There are however, no long-term data on the AIP maintenance phase. Finally, there are no established guidelines or evidence to confirm adequacy in the protein intake.
4.4. Expanding the AIP role
Aside from incorporating the AIP into the management of autoimmune diseases, several other conditions may benefit from its application: individuals experiencing unexplained symptoms such as eczema and migraines, those seeking to gain a deeper understanding of their body, individuals aiming to enhance gut health and overall well-being, those looking to identify food allergies, sensitivities and intolerances, individuals with eosinophilic esophagitis [54], and people diagnosed with attention deficit hyperactivity disorder (ADHD) [55]. Despite the low evidence, several societies present AIP as a possible diet regimen for patients with autoimmune diseases like systemic lupus erythematosus [56].
5. Conclusion
Despite the limited clinical evidence, the AIP has been gaining popularity recently. It consists of a personalized elimination diet, with the rationale of reducing inflammation and alleviating symptoms associated with autoimmune diseases. In parallel, it promotes nutrient-rich, whole foods supporting gut health and supplying essential vitamins and minerals for immune and intestinal function and overall well-being. Although the AIP can be challenging, it is a customized and personalized diet plan that suits the patients’ individual needs, being a promising, powerful diagnostic and therapeutic tool.
Horizontal elimination diets like the FODMAP and the Paleo lack consistency in favorable outcomes across different populations [57]. On the other hand, the AIP has the potential to identify individual immune responses and tailor intake in a personalized approach. In this manner, it aligns with the contemporary call for customized nutrition. Whether the AIP will be the holy grail or the next fad diet for autoimmune diseases will depend on the research quantity and quality. Controlling autoimmunity with diet would be life-changing for patients. For this, a pledge for high-quality studies is made to elucidate the biological mechanisms of the AIP and expand the knowledge of its efficacy and safety.
CRediT authorship contribution statement
Eleni C. Pardali: Writing – original draft, Visualization, Conceptualization. Arriana Gkouvi: Writing – original draft, Visualization, Investigation, Data curation. Kalliopi K. Gkouskou: Writing – review & editing. Anastasios C. Manolakis: Writing – review & editing, Investigation. Christina Tsigalou: Writing – review & editing, Project administration, Funding acquisition. Dimitrios G. Goulis: Writing – review & editing, Supervision, Methodology. Dimitrios P. Bogdanos: Writing – review & editing, Supervision, Methodology, Conceptualization. Maria G. Grammatikopoulou: Writing – review & editing, Writing – original draft, Supervision, Methodology, Data curation, Conceptualization.
Funding
This review received no external funding.
Conflicts of interest
None.
References
- 1.Szekanecz Z., McInnes I.B., Schett G., Szamosi S., Benkő S., Szűcs G. Autoinflammation and autoimmunity across rheumatic and musculoskeletal diseases. Nat Rev Rheumatol. 2021;17:585–595. doi: 10.1038/S41584-021-00652-9. [DOI] [PubMed] [Google Scholar]
- 2.Zhao Q. Bispecific antibodies for autoimmune and inflammatory diseases: clinical progress to date. BioDrugs. 2020;34:111–119. doi: 10.1007/s40259-019-00400-2. [DOI] [PubMed] [Google Scholar]
- 3.Chen J.Q., Szodoray P., Zeher M. Toll-like receptor pathways in autoimmune diseases. Clin Rev Allergy Immunol. 2016;50:1–17. doi: 10.1007/S12016-015-8473-Z. [DOI] [PubMed] [Google Scholar]
- 4.Miller F.W. The increasing prevalence of autoimmunity and autoimmune diseases: an urgent call to action for improved understanding, diagnosis, treatment, and prevention. Curr Opin Immunol. 2023;80 doi: 10.1016/J.COI.2022.102266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhang F., Cui Y., Gao X. Time trends in the burden of autoimmune diseases across the BRICS: an age–period–cohort analysis for the GBD 2019. RMD Open. 2023;9 doi: 10.1136/RMDOPEN-2023-003650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Conrad N., Misra S., Verbakel J.Y., Verbeke G., Molenberghs G., Taylor P.N., et al. Incidence, prevalence, and co-occurrence of autoimmune disorders over time and by age, sex, and socioeconomic status: a population-based cohort study of 22 million individuals in the UK. Lancet. 2023;401:1878–1890. doi: 10.1016/S0140-6736(23)00457-9. [DOI] [PubMed] [Google Scholar]
- 7.Snetselaar L.G., Cheek J.J., Fox S.S., Healy H.S., Schweizer M.L., Bao W., et al. Efficacy of diet on fatigue and quality of life in multiple sclerosis. Neurology. 2023;100:e357–e366. doi: 10.1212/WNL.0000000000201371. [DOI] [PubMed] [Google Scholar]
- 8.Grammatikopoulou M.G., Marakis G., Gkiouras K., Athanatou D., Maraki M.I., Bogdanos D.P. Fly me to the immune: immunonutrition in rheumatic diseases. Mediterr J Rheumatol. 2023;34:30–36. doi: 10.31138/MJR.34.1.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Manzel A., Muller D.N., Hafler D.A., Erdman S.E., Linker R.A., Kleinewietfeld M. Role of “western diet” in inflammatory autoimmune diseases compliance with ethics guidelines human and animal rights and informed consent. Curr Allergy Asthma Rep. 2014;14:404. doi: 10.1007/s11882-013-0404-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Thorburn A.N., Macia L., Mackay C.R. Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity. 2014;40:833–842. doi: 10.1016/J.IMMUNI.2014.05.014. [DOI] [PubMed] [Google Scholar]
- 11.Ballantyne S. Victory Belt Publishing; 2013. The Paleo Approach : reverse autoimmune disease and heal your body. Las Vegas. [Google Scholar]
- 12.Akdis C.A. Does the epithelial barrier hypothesis explain the increase in allergy, autoimmunity and other chronic conditions? Nat Rev Immunol. 2021;21:739–751. doi: 10.1038/s41577-021-00538-7. [DOI] [PubMed] [Google Scholar]
- 13.Christovich A., Luo X.M. Gut microbiota, leaky gut, and autoimmune diseases. Front Immunol. 2022;13 doi: 10.3389/FIMMU.2022.946248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mu Q., Kirby J., Reilly C.M., Luo X.M. Leaky gut as a danger signal for autoimmune diseases. Front Immunol. 2017;8:598. doi: 10.3389/fimmu.2017.00598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Di Vincenzo F., Del Gaudio A., Petito V., Lopetuso L.R., Scaldaferri F. Gut microbiota, intestinal permeability, and systemic inflammation: a narrative review. Intern Emerg Med. 2024;19:275–293. doi: 10.1007/s11739-023-03374-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sundaram T.S., Giromini C., Rebucci R., Pistl J., Bhide M., Baldi A. Role of omega-3 polyunsaturated fatty acids, citrus pectin, and milk-derived exosomes on intestinal barrier integrity and immunity in animals. J Anim Sci Biotechnol. 2022;13:40. doi: 10.1186/s40104-022-00690-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Vancamelbeke M., Vermeire S. The intestinal barrier: a fundamental role in health and disease. Expet Rev Gastroenterol Hepatol. 2017;11:821–834. doi: 10.1080/17474124.2017.1343143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Persson E.K., Scott C.L., Mowat A.M., Agace W.W. Dendritic cell subsets in the intestinal lamina propria: ontogeny and function. Eur J Immunol. 2013;43:3098–3107. doi: 10.1002/eji.201343740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kim E., Tran M., Sun Y., Huh J.R. Isolation and analyses of lamina propria lymphocytes from mouse intestines. STAR Protoc. 2022;3 doi: 10.1016/j.xpro.2022.101366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Karczewski J., Poniedziałek B., Adamski Z., Rzymski P. The effects of the microbiota on the host immune system. Autoimmunity. 2014;47:494–504. doi: 10.3109/08916934.2014.938322. [DOI] [PubMed] [Google Scholar]
- 21.Turner M.D., Nedjai B., Hurst T., Pennington D.J. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta. 2014;1843:2563–2582. doi: 10.1016/j.bbamcr.2014.05.014. [DOI] [PubMed] [Google Scholar]
- 22.Thoo L., Noti M., Krebs P. Keep calm: the intestinal barrier at the interface of peace and war. Cell Death Dis. 2019;10:849. doi: 10.1038/s41419-019-2086-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nicholson L., Raveney B., Munder M. Monocyte dependent regulation of autoimmune inflammation. Curr Mol Med. 2009;9:23–29. doi: 10.2174/156652409787314499. [DOI] [PubMed] [Google Scholar]
- 24.Arroyo Hornero R., Hamad I., Côrte-Real B., Kleinewietfeld M. The Impact of dietary components on regulatory T cells and disease. Front Immunol. 2020;11:253. doi: 10.3389/FIMMU.2020.00253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Fasano A., Sapone A., Zevallos V., Schuppan D. Nonceliac gluten sensitivity. Gastroenterology. 2015;148:1195–1204. doi: 10.1053/J.GASTRO.2014.12.049. [DOI] [PubMed] [Google Scholar]
- 26.Wilders-Truschnig M., Mangge H., Lieners C., Gruber H.J., Mayer C., März W. IgG antibodies against food antigens are correlated with inflammation and intima media thickness in obese juveniles. Exp Clin Endocrinol Diabetes. 2008;116:241–245. doi: 10.1055/S-2007-993165. [DOI] [PubMed] [Google Scholar]
- 27.Mankarious S., Lee M., Fischer S., Pyun K.H., Ochs H.D., Oxelius V.A., et al. The half-lives of IgG subclasses and specific antibodies in patients with primary immunodeficiency who are receiving intravenously administered immunoglobulin. J Lab Clin Med. 1988;112:634–640. [PubMed] [Google Scholar]
- 28.Jew S., Abumweis S.S., Jones P.J.H. Evolution of the human diet: linking our ancestral diet to modern functional foods as a means of chronic disease prevention. J Med Food. 2009;12:925–934. doi: 10.1089/JMF.2008.0268. [DOI] [PubMed] [Google Scholar]
- 29.DeLa O.V., Zazpe I., Martínez J.A., Santiago S., Carlos S., Zulet M.Á., et al. Scoping review of Paleolithic dietary patterns: a definition proposal. Nutr Res Rev. 2021;34:78–106. doi: 10.1017/S0954422420000153. [DOI] [PubMed] [Google Scholar]
- 30.Boers I., Muskiet F.A., Berkelaar E., Schut E., Penders R., Hoenderdos K., et al. Favourable effects of consuming a Palaeolithic-type diet on characteristics of the metabolic syndrome: a randomized controlled pilot-study. Lipids Health Dis. 2014;13:160. doi: 10.1186/1476-511X-13-160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lindeberg S., Jönsson T., Granfeldt Y., Borgstrand E., Soffman J., Sjöström K., et al. A Palaeolithic diet improves glucose tolerance more than a Mediterranean-like diet in individuals with ischaemic heart disease. Diabetologia. 2007;50:1795–1807. doi: 10.1007/S00125-007-0716-Y/TABLES/7. [DOI] [PubMed] [Google Scholar]
- 32.Shah S., Mahamat-Saleh Y., Hajji-Louati M., Correia E., Oulhote Y., Boutron-Ruault M.C., et al. Palaeolithic diet score and risk of breast cancer among postmenopausal women overall and by hormone receptor and histologic subtypes. Eur J Clin Nutr. 2023;77:596–602. doi: 10.1038/s41430-023-01267-x. [DOI] [PubMed] [Google Scholar]
- 33.Frassetto L.A., Schloetter M., Mietus-Synder M., Morris R.C., Sebastian A. Metabolic and physiologic improvements from consuming a paleolithic, hunter-gatherer type diet. Eur J Clin Nutr. 2009;63:947–955. doi: 10.1038/ejcn.2009.4. [DOI] [PubMed] [Google Scholar]
- 34.Otten J., Andersson J., Stáhl J., Stomby A., Saleh A., Waling M., et al. Exercise training adds cardiometabolic benefits of a paleolithic diet in type 2 diabetes mellitus. J Am Heart Assoc. 2019;8 doi: 10.1161/JAHA.118.010634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Masharani U., Sherchan P., Schloetter M., Stratford S., Xiao A., Sebastian A., et al. Metabolic and physiologic effects from consuming a hunter-gatherer (Paleolithic)-type diet in type 2 diabetes. Eur J Clin Nutr. 2015;69:944–948. doi: 10.1038/EJCN.2015.39. [DOI] [PubMed] [Google Scholar]
- 36.Frączek B., Pięta A., Burda A., Mazur-Kurach P., Tyrała F. Paleolithic diet—effect on the health status and performance of athletes? Nutrients. 2021;13:1019. doi: 10.3390/NU13031019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gibson P.R., Shepherd S.J. Evidence-based dietary management of functional gastrointestinal symptoms: the FODMAP approach. J Gastroenterol Hepatol. 2010;25:252–258. doi: 10.1111/J.1440-1746.2009.06149.X. [DOI] [PubMed] [Google Scholar]
- 38.Høst A. Cow's milk allergy. J R Soc Med. 1997;90:34–39. doi: 10.1177/0141076897090030S06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Spitler D.K. Elimination diets and patient's allergies. A handbook of allergy. Bull Med Libr Assoc. 1944;32:534. [Google Scholar]
- 40.Holmes G.K.T., Prior P., Lane M.R., Pope D., Allan R.N. Malignancy in coeliac disease--effect of a gluten free diet. Gut. 1989;30:333–338. doi: 10.1136/GUT.30.3.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gibson P.R., Shepherd S.J. Personal view: food for thought--western lifestyle and susceptibility to Crohn's disease. The FODMAP hypothesis. Aliment Pharmacol Ther. 2005;21:1399–1409. doi: 10.1111/J.1365-2036.2005.02506.X. [DOI] [PubMed] [Google Scholar]
- 42.Bisht B., Darling W.G., Grossmann R.E., Shivapour E.T., Lutgendorf S.K., Snetselaar L.G., et al. A multimodal intervention for patients with secondary progressive multiple sclerosis: feasibility and effect on fatigue. J Alternative Compl Med. 2014;20:347–355. doi: 10.1089/ACM.2013.0188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ihnatowicz P., Gębski J., Drywień M.E. Effects of Autoimmune Protocol (AIP) diet on changes in thyroid parameters in Hashimoto's disease. Ann Agric Environ Med. 2023;30:513–521. doi: 10.26444/AAEM/166263. [DOI] [PubMed] [Google Scholar]
- 44.Abbott R.D., Sadowski A., Alt A.G. Efficacy of the autoimmune protocol diet as part of a multi-disciplinary, supported lifestyle intervention for Hashimoto's thyroiditis. Cureus. 2019;11:e4556. doi: 10.7759/CUREUS.4556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chandrasekaran A., Groven S., Lewis J.D., Levy S.S., Diamant C., Singh E., et al. An autoimmune protocol diet improves patient-reported quality of life in inflammatory bowel disease. Crohn’s Colitis. 2019;360 doi: 10.1093/CROCOL/OTZ019. 1:otz01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Konijeti G.G., Kim N., Lewis J.D., Groven S., Chandrasekaran A., Grandhe S., et al. Efficacy of the autoimmune protocol diet for inflammatory bowel disease. Inflamm Bowel Dis. 2017;23:2054–2060. doi: 10.1097/MIB.0000000000001221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chandrasekaran A., Molparia B., Akhtar E., Wang X., Lewis J.D., Chang J.T., et al. The autoimmune protocol diet modifies intestinal RNA expression in inflammatory bowel disease. Crohn’s Colitis. 2019;360 doi: 10.1093/CROCOL/OTZ016. 1:otz016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mackay S., Jalili-Moghaddan S., Gibbs M., Mcneill J., Zinn C., Mearns G., et al. What is the efficacy of the autoimmune protocol (AIP) diet in people with rheumatoid arthritis? A mixed-methods pilot intervention study. Med Sci Forum. 2023;18:10. doi: 10.3390/MSF2023018010. [DOI] [Google Scholar]
- 49.McNeill J. A pilot study to test the effect of the autoimmune protocol l diet in people with rheumatoid arthritisdiet in people with rheumatoid arthritis. Rangahau Aranga AUT Grad Rev. 2022;1:1. doi: 10.24135/RANGAHAU-ARANGA.V1I1.67. [DOI] [Google Scholar]
- 50.Hurrell R., Egli I. Iron bioavailability and dietary reference values. Am J Clin Nutr. 2010;91 doi: 10.3945/AJCN.2010.28674F. 1461S – 7. [DOI] [PubMed] [Google Scholar]
- 51.Titcomb T.J., Bisht B., Moore D.D., Chhonker Y.S., Murry D.J., Snetselaar L.G., et al. Eating pattern and nutritional risks among people with multiple sclerosis following a modified paleolithic diet. Nutrients. 2020;12:1844. doi: 10.3390/NU12061844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Cashman K.D. Vitamin D deficiency: defining, prevalence, causes, and strategies of addressing. Calcif Tissue Int. 2020;106:14–29. doi: 10.1007/S00223-019-00559-4. [DOI] [PubMed] [Google Scholar]
- 53.Reid I.R. Should we prescribe calcium supplements for osteoporosis prevention? J Bone Metab. 2014;21:21–28. doi: 10.11005/JBM.2014.21.1.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gonsalves N., Yang G.Y., Doerfler B., Ritz S., Ditto A.M., Hirano I. Elimination diet effectively treats eosinophilic esophagitis in adults; food reintroduction identifies causative factors. Gastroenterology. 2012;142 doi: 10.1053/J.GASTRO.2012.03.001. 1451–e15. [DOI] [PubMed] [Google Scholar]
- 55.Pelsser L.M., Frankena K., Toorman J., Savelkoul H.F., Dubois A.E., Pereira R.R., et al. Effects of a restricted elimination diet on the behaviour of children with attention-deficit hyperactivity disorder (INCA study): a randomised controlled trial. Lancet (London, England) 2011;377:494–503. doi: 10.1016/S0140-6736(10)62227-1. [DOI] [PubMed] [Google Scholar]
- 56.Schoenbach A. Autoimmune protocol diet (AIP) for lupus. LupusCorner. 2020 https://lupuscorner.com/autoimmune-protocol-diet-aip-for-lupus/ Accessed on 30th Dec 2024. [Google Scholar]
- 57.Grammatikopoulou M.G., Goulis D.G., Gkiouras K., Nigdelis M.P., Papageorgiou S.T., Papamitsou T., et al. Low FODMAP diet for functional gastrointestinal symptoms in quiescent inflammatory bowel disease: a systematic review of randomized controlled trials. Nutrients. 2020;12:3648. doi: 10.3390/NU12123648. [DOI] [PMC free article] [PubMed] [Google Scholar]