Basic Study Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Pharmacol Ther. Nov 8, 2020; 11(5): 79-92
Published online Nov 8, 2020. doi: 10.4292/wjgpt.v11.i5.79
Oral encapsulated transforming growth factor β1 reduces endogenous levels: Effect on inflammatory bowel disease
Laura Hammer, Stacia Furtado, Dominick L Auci, Department of Research and Development, TherapyX, Buffalo, NY 14214, United States
Stacia Furtado, Edith Mathiowitz, Department of Molecular Pharmacology, Brown University, Providence, RI 02912, United States
ORCID number: Laura Hammer (0000-0002-2324-3019); Stacia Furtado (0000-0001-7079-4617); Edith Mathiowitz (0000-0003-3054-7130); Dominick L Auci (0000-0001-5315-9873).
Author contributions: Auci DL and Hammer L designed and coordinated the studies, analyzed data and wrote the manuscript; Hammer L performed in life phases and serum and tissue analysis; Furtado S, and Mathiowitz E prepared drug products and reviewed the manuscript.
Supported by National Institute of Allergy and Infectious Diseases of the National Institutes of Health under award, No. 5R44AI080009.
Institutional animal care and use committee statement: All experiments were conducted in accordance with policies of the NIH Guide for the Care and Use of Laboratory Animals and Institutional Animal Care and Use Committee (IACUC) of the State University of New York at Buffalo, or Comparative Biosciences. Approved protocol MIC24125Y.
Conflict-of-interest statement: Dr. Auci reports grants from NIH, during the conduct of the study; In addition, Dr. Auci has a patent Micronized freeze-dried particles issued, and a patent Compositions for stabilizing and delivering proteins pending and Authors hold equity in Therapyx.
Data sharing statement: No additional data are available.
ARRIVE guidelines statement: The authors have read the ARRIVE guidelines, and the manuscript was prepared and revised according to the ARRIVE guidelines.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Dominick L Auci, PhD, Senior Researcher, Department of Research and Development, TherapyX, 108 Biomedical Research Building, 3435 Main Street, Buffalo, NY 14214, United States. dauci@therapyxinc.com
Received: May 7, 2020
Peer-review started: May 7, 2020
First decision: June 7, 2020
Revised: June 18, 2020
Accepted: October 9, 2020
Article in press: October 9, 2020
Published online: November 8, 2020

Abstract
BACKGROUND

TreXTAM® is a combination of the key regulatory cytokine transforming growth factor beta (TGFβ) and all trans retinoic acid (ATRA) microencapsulated for oral delivery to immune structures of the gut. It is in development as a novel treatment for inflammatory bowel disease (IBD).

AIM

To measure TGFβ levels in blood and tissue after oral administration of encapsulated TGFβ.

METHODS

Animals were orally administered encapsulated TGFβ by gavage. Levels of drug substance in blood and in gut tissues at various times after administration were measured by ELISA.

RESULTS

We made the surprising discovery that oral administration of TreXTAM dramatically (approximately 50%) and significantly (P = 0.025) reduced TGFβ levels in colon, but not small intestine or mesenteric lymph nodes. Similarly, levels in rat serum after 25 d of thrice weekly dosing with either TreXTAM, or microencapsulated TGFβ alone (denoted as TPX6001) were significantly (P < 0.01) reduced from baseline levels. When tested in the SCID mouse CD4+CD25- adoptive cell transfer (ACT) model of IBD, oral TPX6001 alone provided only a transient benefit in terms of reduced weight loss.

CONCLUSION

These observations suggest a negative feedback mechanism in the gut whereby local delivery of TGFβ results in reduced local and systemic levels of the active form of TGFβ. Our findings suggest potential clinical implications for use of encapsulated TGFβ, perhaps in the context of IBD and/or other instances of fibrosis and/or pathological TGFβ signaling.

Key Words: Transforming growth factor beta, All trans retinoic acid, Ulcerative colitis, Crohn’s disease, Inflammatory bowel disease, Regulatory T cells

Core Tip: The observations suggest a negative feedback mechanism in the gut whereby local delivery of transforming growth factor beta (TGFβ) to immune structures of the gut results in reduced local and systemic levels of the active form of TGFβ.



INTRODUCTION

TreXTAM® is a proprietary micro-encapsulated drug product in development as an oral treatment for inflammatory bowel disease (IBD). It is the combination of the key regulatory cytokine transforming growth factor beta (TGFβ) encapsulated in into poly-lactic acid (PLA) particles; along with a signaling form of vitamin A, all trans retinoic acid (ATRA), encapsulated in poly D,L-lactide-co-glycolide (PLGA) particles[1]. Simultaneous ATRA and TGFβ signals synergize in promoting the differentiation and stabilization of regulatory T cells[2]. This is a completely novel strategy for the treatment of IBD, as no similar products exist. However, unlike ATRA, TGFβ is a protein macromolecule that must be protected against hydrolysis in the stomach to be effective via the oral route[3].

Encapsulation remains one of the most promising methods to protect drug substances and to achieve local, sustained release. Efforts have generally focused on siRNA[4], small molecules[5], peptides[6] and cytokines[7] and involve polymer encapsulation accomplished via combinations of phase separation or precipitation, emulsion/solvent evaporation[8-15] and/or spraying methods[16-20]. However, loss of bioactivity during manufacturing, poorly controlled release rates, and difficulties with large-scale production of accurately sized particles are some of the formidable challenges preventing commercialization.

To address these challenges, we pioneered the development of phase inversion nano-encapsulation (PIN®) technology that utilizes a non-mechanical approach to preserve the structural integrity of macromolecules during the drug product manufacturing process. PIN encapsulated cytokines have demonstrated stability, bioactivity and efficacy in various preclinical models[21-26]. Particles with an average diameter of 0.1-5 microns[6], are ideally suited to oral delivery as particles smaller than 5 microns in diameter readily traverse the gastrointestinal barrier[27-29]. Indeed, we had previously shown that orally administered insulin encapsulated in PIN particles resulted in localization of drug product to the gut, and efficient uptake at the intestinal border[6,7].

More recently we applied PIN technology to the development of TreXTAM and showed that oral administration effectively ameliorated disease in two different rodent IBD models[1]. Broadly, treatment of mice with established disease using the optimized dose/frequency regimen, achieved a dramatic 2 to 9-fold reduction in multiple markers of disease compared to control groups within 2 wk, in some cases approaching normal values. Importantly, treatment enhanced long-term survival over eight weeks with no detectable toxicity. Activity was associated with enhanced Foxp3 expression in the colonic lamina propria CD4+ CD25+ T-cells, and required both TGFβ and ATRA for maximal efficacy. We have recently reviewed potential cellular and molecular mechanisms driving synergy, including cross-talk between ATRA and TGFβ signal transduction pathways[2].

During TreXTAM development, we studied TGFβ pharmacokinetics after oral administration of TreXTAM, or after the encapsulated cytokine (TPX6001) was given alone, without ATRA. We made the surprising discovery that oral administration of TreXTAM dramatically reduced TGFβ levels in colon and in blood, to below baseline levels. When encapsulated TGFβ (TPX6001) was given alone, three times a week for 25 d, we likewise observed serum TGFβ decreases below baseline (untreated) levels. Oral treatment with TPX6001 alone transiently ameliorated weight loss in the murine adoptive cell transfer (ACT) model of IBD. These observations suggest a negative feedback mechanism in the gut whereby local delivery of TGFβ results in reduced local and systemic levels of the active form of TGFβ. This finding suggests potential clinical implications for use of encapsulated TGFβ in the context of IBD and/or pathological TGFβ signaling.

MATERIALS AND METHODS
Preparation and characterization of TGFβ and ATRA loaded formulations

Microsphere preparation: For tissue studies, TGFβ (Peprotech, Rocky Hill, NJ, United States) was encapsulated into bench-top scale, poly-lactic acid (PLA) particles (0.285 mg TGFβ per gram of final drug product; for simplicity and clarity the abbreviation TGFβ refers specifically to TGFβ1, unless otherwise noted) using PIN as described previously[30]. ATRA (Sigma) was encapsulated into poly-lactic-co-glycolic acid (PLGA) particles (1 mg of ATRA per gram of particles) using a modification of the solvent evaporation technique as in previous studies[31].

For PK studies, TGFβ and ATRA loaded microspheres (denoted TPX6001 and TPX7001, respectively) were synthesized at Lonza-Bend, Bend Oregon using a proprietary two-step spray dry process to manufacture larger, scaled up quantities. Briefly, in Step 1, lyophilized protein is mixed with excipients and dispersed. In Step 2, micronized protein + excipients are encapsulated, precipitated and collected. To reduce dose mass, TGFβ and ATRA spray dry drug products were loaded at 1 mg/g and 2 mg/g w/v. The release kinetics, bioactivity, morphology, long-term (1 year) stability, as well as the physicochemical properties of glass transition temperature and crystallinity were essentially identical in the bench lots and spray dried particles (data not shown).

TGFβ and ATRA loaded PLA and PLGA particles were mixed cage-side in the indicated proportions to create TreXTAM, a proprietary combinatorial product designed to provide both TGFβ and ATRA signals thought to drive the development of regulatory T cells[32-35].

In vitro drug substance release: Formulations were release-tested using an in vitro release assay described previously[24]. Briefly, for TGFβ, 0.2 mL of a 10 mg/mL particle suspension was transferred to the wells of a 96-well plate in triplicate. The plate was incubated at 37 oC in 5% CO2, the supernatants were sampled at the indicated time points and stored at -20 oC until use. ATRA was extracted and measured by HPLC as in our previous studies[36]. The immune-reactive, active form of TGFβ was measured by assaying non-acidified samples in an ELISA (R&D Systems Quantikine ELISA kit Catalog# MB100B). This assay does not have significant cross-reactivity or interference with TGFβ2 or TGFβ3, and does not detect the latent form of TGFβ1 without acid treatment. ATRA extraction and analysis was performed as follows: 10 ± 0.1 mg ATRA containing microspheres were weighed into 15 mL Falcon tubes for each terminal time-point. 1 mL of 1 × PBS was added and placed on end-over-end rotator at 37 ºC. At predetermined time-points, the tubes were centrifuged and supernatant discarded. The remaining microspheres were flash frozen and lyophilized for 24 h. Microsphere samples were then extracted by adding 5 mL of pH 7 mobile phase (68:24:8 ratio of acetonitrile: 1% glacial acetic acid:ethanol) and bath sonicating for 45 min. Extracted samples were then run on a HPLC using a Waters Symmetry C18 Column (5.0 µm, 3.9 mm × 150 mm) at a flow rate of 1 mL/min using pH 7 mobile phase. Absorbance was measured at 356 nm.

Pharmacokinetic studies

Animals: The in-life phase of these studies was performed at Comparative Biosciences, Sunnyvale, California. 7 to 9-wk old Sprague-Dawley rats (males and females) were kept under standard laboratory conditions with free access to food and water. They were allowed to adapt one week before starting the study. The care and use of laboratory animals was in accordance with relevant IACUC-approved animal use protocols.

Administration of encapsulated drug products: A 0.5-mL aliquot of TreXTAM (or TPX6001 alone) in aqueous suspension was prepared by reconstitution of drug products (TPX7001 and/or TPX6001) with distilled water and mixed in appropriate w/v proportions to achieve the targeted dosing. Animals were dosed by oral gavage. Blood samples were collected at fixed times after dosing.

Tissues analysis: 7 to 9-wk old male Sprague-Dawley rats n = 3 per group) were untreated, or treated (oral gavage) with TreXTAM three times per week for four weeks. Four hours after the final dose, gut tissues were taken, frozen at -20 oC and stored until used. Tissues were then thawed and homogenized using a glass tube with the pestle insert, in the presence of EDTA-free SIGMAFAST™ Protease Inhibitor Cocktail Tablets (Sigma-Aldrich) used as per manufacturer’s instructions. Levels of TGFβ1 and ATRA in lysates were measured as described above.

Serum analysis: Serum levels of TGFβ1 were measured using an ELISA kit (R&D Systems, Minneapolis, MN; see above) with a slight modification from manufacturer’s instructions. Samples were not acid- activated, minimizing detection of endogenous latent cytokine. For ATRA, a high-performance liquid chromatograph combined with a triple quadrupole mass spectrometer was used as in our previous studies[36].

SCID mouse CD4+CD25- T cell transfer colitis model

The model was chosen because it recapitulates a regulatory T cell immunological basis of colitis, and was performed as in our previous studies[1]. Briefly:

Animals: Six to 8-wk old BALB/c and CB-17 SCID mice (males and females; Jackson Laboratories, Bar Harbor, MA, United States) were kept under standard laboratory conditions with free access to food and water and allowed to adapt one week before starting the study. The care and use of laboratory animals was in accordance with a University at Buffalo IACUC-approved animal use protocol.

Isolation of CD4+CD25- T cells: CD4+ CD25- T-cells were purified from the spleens of naïve BALB/c mice by magnetic bead separation using MACS® column and separator according to manufacturer’s instructions (Miltenyi Biotech, San Diego, CA, United States). Purity and viability (> 95%) were assessed by flow cytometry (FACScan, Becton Dickinson, San Jose, CA, United States).

Induction of colitis: Purified CD4+CD25- T-cells were adoptively-transferred to SCID recipients (4 × 105 cells per mouse, i.p.). Mice were randomized into groups when 10% of mice show 5% or greater weight loss and/or soft or bloody stools and treatment (3 × per week via oral gavage) started. Daily disease score was recorded for each animal as in our previous studies[1] and summarized for each group as cumulative disease score during treatment. Last recorded values of animals that died during treatment were brought forward. At the end of the treatment period, all mice were sacrificed, and colons scored grossly for pathology on a 0 (normal) to 5 (diseased; elongated, inflamed, lacking definable stools) scale. Histology was also performed as in our previous studies[1]. Six to eight H&E sections of colon representing ascending, transverse and descending colon per mouse were evaluated independently, in blinded fashion, by a board-certified pathologist (Pacific Tox Path, LLC, Ellensburg, WA, United States). A composite inflammation score was calculated based on (0-3) severity and extent of cellular infiltration, amount of mucus and degree of proliferation (maximum score of 12).

Statistical analysis

Significance (P ≤ 0.05) between experimental and control groups was determined using Student’s t-test analysis. In experiments with multiple groups, homogeneity of inter-group variance was analyzed by ANOVA.

RESULTS
In vitro release patterns of TGFβ and ATRA and typical appearance of PLA and PLGA microsphere particles

For tissue studies, and for ACT studies, TGFβ and ATRA were encapsulated using PIN or solvent evaporation techniques bench top-processes (respectively) at 0.285 mg/g and 1 mg/g w/w, respectively. At 24 h, both TGFβ and ATRA drug products released bioactive (confirmed using TGFβ sensitive mouse lymphoblast cell line HT-2 or ATRA sensitive murine melanoma B16-F1cells; data not shown) TGFβ or ATRA (Figure 1A and B respectively) as expected, indicating that both drug substances could potentially be delivered in active forms, simultaneously, in vivo after oral administration.

Figure 1
Figure 1 Release profiles of transforming growth factor -loaded poly-lactic acid microspheres and all trans retinoic acid-loaded poly-lactic-co-glycolic acid microspheres. Transforming growth factor (TGF) was encapsulated in poly-lactic acid (PLA) microspheres (285 μg of TGF per gram of particles) using Phase Inversion Nano-encapsulation (PIN). All trans retinoic acid (ATRA) was encapsulated into poly-lactic-co-glycolic acid (PLGA) microspheres (1 mg of ATRA per gram of particles) using a modification of the solvent evaporation technique (see methods section). A: TGF-loaded microspheres were release-tested using the in vitro release assay as described in the methods section; B: ATRA-loaded microspheres were release-tested using an in vitro extraction assay as described in the methods section. Data are expressed as pg/mL or as μg/mL ± SE. TGFβ: Transforming growth factor ; ATRA: All trans retinoic acid.
TGFβ and ATRA in small and large intestine and MLN after oral Administration of TreXTAM to male rats

To assess delivery of ATRA and TGFβ to gut, male Sprague-Dawley rats were fed with either blank particles or with TreXTAM (60 mg/kg and 30 mg/kg of TGFβ and ATRA loaded particles; denoted TPX6001 and TPX7001, respectively, and loaded at 0.286 mg/g and 1 mg/g, approximately 17 and 30 μg/kg respectively) three times per week for four weeks. Four hours after the final treatment, small intestine, large intestine and MLN were collected from each animal and frozen at -20 oC. ATRA and TGFβ levels in small and large intestine, as well as MLN, were determined by HPLC or ELISA, respectively (limit of detection 0.75 ng/mL and 0.4 pg/TGFβ/100 μg of protein, respectively). Levels of ATRA in small intestine and MLN of treated and untreated animals were at the limit of detection. Levels of ATRA in colon were virtually the same in treated and untreated animals (data not shown). TGFβ was also negligible in small intestine and MLN of treated and untreated animals. However, TGFβ levels in colon of treated animals were decreased over 50% compared to untreated animals (Figure 2). This difference was significant (P = 0.025) suggesting a treatment associated attenuation of endogenous active TGFβ in colon tissue. Since those initial studies, we scaled up production of PLA encapsulated TGFβ (TPX6001) using the proprietary two-step stray dried manufacturing process described in the methods section. Production of PLGA encapsulated ATRA (TPX7001) has also been scaled up using spray drying methods. Release rates and physiochemical properties of the spray dried and bench top materials were virtually identical (data not shown). All pharmacokinetic work to follow was performed using spray-dried TGFβ PLA and ATRA PLGA (loaded at 0.1 and 0.2%, respectively) material.

Figure 2
Figure 2 Effect of oral treatment with TreXTAM on levels of transforming growth factor in colon. Treated rats (n =3) received TreXTAM [60 mg/kg encapsulated transforming growth factor (TGF) and 30 mg/kg encapsulated all trans retinoic acid (ATRA)] three times a week for four weeks. Colons of these animals were taken 4 h after the final dose, along with tissues from age and sex matched untreated animals. All tissues were frozen at -20 oC and stored until used. Tissues were then thawed and homogenized using a glass tube with the pestle insert, in the presence of EDTA-free SIGMAFAST™ Protease Inhibitor Cocktail Tablets (used as per manufacturer’s instructions). Levels of TGF in lysates were determined by ELISA according to manufacturer’s instructions but without acid activation. (Quantikine, R&D Systems, Minneapolis, MN, United States). Total protein concentration was determined by BCA Protein Assay- Pierce (Thermo Fisher Cat# 23227) Data are expressed as pg/100 μg protein ± standard deviation. TGFβ: Transforming growth factor .
Pharmacokinetics following oral administration of TreXTAM

We could not directly demonstrate simultaneous delivery of TGFβ or ATRA to gut tissue by oral TreXTAM (although we could see biological effects[1]). To further investigate this issue in vivo, and as part of our development efforts, we tested oral TreXTAM in a 28-d GLP rat toxicology study. The relevant pharmacokinetic for ATRA after TreXTAM administration has been published previously[36]. Those studies reported that after a single oral TreXAM administration, serum ATRA levels peaked with a Tmax of 60 min and t ½ of 143 min.

We report here that after oral administration of TreXTAM (30 mg/kg spray-dried encapsulated TGFβ and 30 mg/kg PLGA encapsulated ATRA) three times a week for 25 d, serum TGFβ levels were significantly reduced compared to those observed in the same animals on day 0, prior to any TreXTAM dosing (Figure 3; NB: The level of ELISA detection is approximately 150 pg/mL). This finding was reminiscent of our observations of reduced TGFβ in colon after dosing (Figure 2). We also note that in the pre- dose, naïve animals n = 24 per sex) females had higher endogenous levels of TGFβ compared to males (492 ± 107 pg/mL vs 324 ± 14 pg/mL). This difference was highly significant (P < 0.0001). Similar observations were reported previously by Knabbe et al[37].

Figure 3
Figure 3 Baseline serum levels of transforming growth factor in naïve and TreXTAM treated animals. Blood was taken from naïve male and female Sprague-Dawley rats (males and females, 3 per sex) before and after treatment with TreXTAM [30 mg/kg encapsulated transforming growth factor (TGF) and 30 mg/kg encapsulated all trans retinoic acid (ATRA)]. Animals were dosed by gavage 3 × per week for 25 d. Serum levels of TGFβ were determined by ELISA without acid activation (R&D Systems Quantikine ELISA Catalog# MB100B). TGFβ: Transforming growth factor .
Oral treatments with PLA encapsulated TGFβ reduce serum levels of TGFβ

We also tested spray-dried PLA encapsulated TGFβ (TPX6001; loaded at 1 mg/g w/w) given alone in a similar 28-d GLP rat toxicology study (Figure 4). Once again, when similar analyses were performed on naive animals and on the same animals that had been dosed three times per week for 25 d, a dramatic and highly significant (P < 0.01) treatment-related reduction in serum TGFβ levels was evident for all dose groups (Figure 4). Indeed, the reduction in baseline serum TGFβ was dose dependent, in that the difference between the low and high dose group was also significant (P < 0.03). It was also interesting to once again note that in naïve pre-dose animals n = 24 per sex), females had significantly (P = 0.001) higher levels of TGFβ than males, (492 ± 107 pg/mL vs 324 ± 14 pg/mL) indicating the same gender bias.

Figure 4
Figure 4 Baseline serum levels of transforming growth factor in naïve rats and in poly-lactic acid encapsulated transforming growth factor treated rats. Blood was taken from naïve Sprague-Dawley rats (males and females, 6 per sex) before and after treatment with poly-lactic acid encapsulated transforming growth factor (TGF). Animals were treated by gavage at doses of 5, 15 or 30 mg/kg, 3 × per week for 25 d. Serum levels of TGFβ were determined by ELISA without acid activation (R&D Systems Quantikine ELISA Catalog# MB100B). Statistical significance (P ≤ 0.02) vs day 25 at 5 mg/kg vs day 25 at 30 mg/kg. All differences between day 0 pre-dose and day 25 pre-dose were significant (P ≤ 0.01). TGFβ: Transforming growth factor .
Effect of TPX6001 on disease in the SCID mouse CD4+CD25- ACT model of IBD

We next tested the IBD therapeutic potential of TPX6001 oral treatments when given alone, without ATRA, in the SCID mouse ACT model of IBD. This single, preliminary study used a highly challenging therapeutic iteration of the model. Treatments began at disease onset. There were no significant differences between groups in terms of body weight or disease score at the start of treatment. We found that TPX6001 treatment resulted in significant attenuation of weight loss (Figure 5). The differences between the 5 mg and 40 mg doses (days 3 to 12) were significant (P = 0. 01) compared to animals treated with blank microspheres. The difference between the 10 mg and blank groups during that same period achieved only a trend (P = 0.12), possibly because of two deaths in the 10 mg group. It is also interesting to note that the high dose group showed the most benefit for the first 7 d of treatment, but then deteriorated rapidly. At the end of the study, for each group, we calculated cumulative disease score during treatment (blank fed group = 52; 5, 10 and 40 mg treatment groups = 49.5, 52.6, and 47, respectively); colon weight to length ratios (blank fed group = 55.3 ± 14.3; healthy age and sex matched controls = 27.9 ± 4.4; 5, 10 and 40 mg treatment groups = 52.8 ± 9.1, 56.5. ± 15.3, and 57.4 ± 9.9, respectively), gross pathology (blank fed group = 2.9 ± 0.9; 5, 10 and 40 mg treatment groups = 3.7 ± 1.4, 2.4. ± 1.4, and 3.8 ± 1.1, respectively) and histology composite inflammation scores (blank fed group = 8.75, and 5, 10 and 40 mg treatment groups = 9.4, 7.8, and 8.1, respectively). We found no significant differences, except at the 10 mg dose (P = 0.003), which again, may have been biased by the deaths of 2 animals in that group. We also note a trend in favor of treatment with respect to cumulative disease score, in the high dose group (P = 0.08). Therefore, we conclude only slight, transient benefit of TPX6001 treatment in this iteration of the ACT model.

Figure 5
Figure 5 Therapeutic activity of transforming growth factor β loaded particles (TPX6001) in the SCID mouse adoptive CD4+ CD25- T-cell transfer model of inflammatory bowel disease. Mice (n = 6-9 per group) with established disease were weighed (day 0) and fed transforming growth factor β1 microspheres (5, 10, or 40 mg/mouse), or blank microspheres (40 mg/mouse) in 0.2 ml water 3 times per week for 2 wk. Mice were monitored for overall disease score and weighed 3 times per week for two weeks. Mice were sacrificed 2 d after the last dose, serum taken, colons weighed and measured; and colons samples prepared for histological analysis (five randomly selected sections from each mouse). Data are expressed as % change in body weight relative to day of first treatment. 5 and 40 mg/mouse TPX6001-treated groups were significantly different (P = 0.01 on days 3-12) from animals treated with blank microspheres.
Multiple (28 d) oral treatments (thrice weekly) with either TreXTAM or encapsulated TGFβ were safe and well tolerated at the highest doses tested

For both TreXTAM and PLA encapsulated TGFβ GLP pharmacokinetic studies, full industry standard toxicology analyses, including clinical observations clinical pathology, necropsy, histopathology and ophthalmology, were also performed on both male and female animals. There were no statistically significant differences in body weights or weekly food intake among groups, and no significant organ weight changes. There were no test article-related histopathological or other findings and no fibrosis was observed with even the highest doses at the end of treatment (Day 28) or at the end of a 56-d recovery period (data not shown). Encapsulated TGFβ, when given alone was as safe and as well tolerated as the TreXTAM combination.

DISCUSSION

We report here that oral TreXTAM produced a surprising and dramatic decrease in serum and colonic TGFβ levels. While we could not directly demonstrate simultaneous delivery of both drug substances to gut tissues, our in vitro and pharmacodynamics observations suggest that it was achieved. In animals given either TreXTAM or PLA encapsulated TGFβ (TPX6001) alone 3 times per week for 25 d, we observed dramatically lower serum TGFβ compared to the same animals before dosing. We also found evidence for a transient benefit of oral TPX6001, at least in terms of weight loss attenuation, in the murine adoptive cell transfer (ACT) model of IBD.

TreXTAM is being developed as treatment for Crohn’s disease (CD) and ulcerative colitis (UC). CD and UC are chronic disorders of the GI tract causing significant morbidity for over 1.4 million Americans[38]. An appreciation of common inflammatory pathways led to the joint designation “IBD”. Symptoms include diarrhea, nausea, abdominal pain, weight loss, increased risk for colorectal cancer[39] and can be fatal[40]. Although etiologies are incompletely understood, genetic, immunologic and environmental factors all make significant contributions[38,41]. Human and animal studies implicate abnormal responses to commensal microflora and perturbed local immune homeostasis[38,39,41]. ‘Biologics’, macromolecules that target inflammatory lymphocytes or the cytokines they produce[42] have emerged as a new class of highly effective treatments. However, an estimated 30% of patients will not respond and of those who initially respond, 50% relapse within a year. A more recent review indicates only modest impact on surgical intervention rates[43]. The need for novel, targeted therapies remains acute. TreXTAM aims to address that need by taking advantage of the synergistic effects of ATRA and TGFβ on the differentiation and stabilization of regulatory T cells[2].

TGFβ is a pleiotropic cytokine with multiple effects on many cell types. It is a key regulator of T-cell biology, impacting thymocyte development, differentiation and effector function[44]. On the one hand, complete loss of TGFβ signaling leads to lymphoproliferative autoimmunity[45-47], on the other hand, systemic administration in microgram doses protects in several autoimmune disease models[48-51]. Unfortunately, TGFβ is also associated with serious side effects, including pulmonary fibrosis[52-55], scleroderma[56], chronic GVHD[57] and glomerulonephropathies[58]. To circumvent these toxicities, local delivery via gene therapy has been proposed, but is inconvenient, transitory, imprecise and immunogenic[48,50,59]. There is no means to control signal transcription or translation, dose schedule, release rates or unwanted immune responses. TreXTAM, aims to circumvent this problems by local delivery and reduces systemic exposure of drug substances with the hope of reducing effective doses and toxicities.

Because of the known fibrotic effects of TGFβ, exacerbation of fibrosis in the context of IBD was a serious concern of oral TreXTAM treatment. The results reported here suggest the opposite might be true, especially in colon, where TreXTAM reduced endogenous TGFβ levels. 28-d TreXTAM repeat dosing studies in rats, like the one reported here for encapsulated TGFβ alone, showed no TreXTAM induced fibrosis in any organ including small intestines and colon (data not shown). Further, we tested TreXTAM, both in healthy mice and in SCID animals with CD4+ CD25- induced colitis, for up to 8 wk, and likewise, found no increases in fibrosis in any organ (Auci et al, unpublished observations). Considering the results reported here, oral treatment with TreXTAM, or even treatment with encapsulated TGFβ alone, may be useful to stimulate autocrine negative feedback and reduce TGFβ levels, to prevent IBD associated fibrosis.

Our inability to detect increased TGFβ in small intestine and MLN after TreXTAM treatment may be due to insignificant amounts of TGFβ delivered despite effective particle uptake in the Peyer’s patches and MLN(7). This may relate to the failure of the particles to reach the colon or rapid degradation and/or deactivation of the released TGFβ. Uptake by other tissues, binding to cell surface proteins or other factors, as well as the potential conversion of TGFβ1 to TGFβ2, 3 or its latent form, would have prevented an increase from being detected. Perhaps most surprisingly, we observed a highly significant TreXTAM-associated decrease (approximately 50%) of active TGFβ in the colon. While this may relate to effects of the particles themselves, a more intriguing possibility involves ATRA amelioration of TGFβ expression and signaling[60]. Several studies report ATRA decreases TGFβ levels and/or signaling in various tissues[61-64]. ATRA modification of TGFβ signaling may also help explain the lack of treatment associated fibrosis observed in our previous studies[1]. Reduction of endogenous TGFβ in colon and its simultaneous delivery to immune structures such as Peyer’s patches and MLN may contribute to the TreXTAM-associated benefits in models of IBD. Like observations in colon, decreases in systemic TGFβ were observed when the encapsulated cytokine was delivered with ATRA in the form of TreXTAM, but also when given alone. Therefore, at least the systemic attenuation of TGFβ levels do not require ATRA and can be achieved with just the encapsulated cytokine. The role of TreXTAM in IBD, including its prophylactic and/or therapeutic usefulness for Crohn’s disease and/or colitis, awaits further studies in various models aimed at determining the contribution each component plays in the efficacy observed.

Our finding of higher levels of TGFβ in female vs male rats is reminiscent of other studies in humans[65] and in non-human primates, where TGFβ levels were found to be higher in young females as compared to males. Interestingly, TGFβ levels decreased with age in females, and increased with age in males, suggesting effect of sex hormones[66]. The wide literature describing activities of TGFβ in the context of autoimmunity and infection has already been extensively reviewed[67], and its consideration is beyond the scope of this work. Suffice to say that an important intersection for the cross talk between TGFβ signaling pathways and sex hormones may lie at the generation and stabilization of regulatory T cells.

Reminiscent of our observations with TreXTAM in tissue, we found that oral TPX6001 when given alone, without ATRA, also reduced serum levels of the endogenous cytokine. The mechanism(s) by which oral treatment with encapsulated TGFβ could lead to reduction in systemic and tissue levels remain unknown. They may relate to synthesis or release of mediators by cells, increased uptake and/or deactivation by other tissues[68] and/or effects on pathways specific to immune structures of the gut. TGFβ is synthesized as an inactive precursor, a complex consisting of a TGFβ dimer, the latency-associated protein, and latent TGFβ binding protein[69]. Before TGFβ can exert its biological effects, both must be dissociated. Therefore, our findings may also relate to specific activation/deactivation pathways, which may be controlled by the gut. It is also possible that our findings relate to switching between immunologically (ELISA) distinct isoforms of TGFβ (1, 2 or 3)[70]. The potential biological significance of such switching is unclear.

To our knowledge, we were the first to administer PLA encapsulated TGFβ via the oral route[1]. Our preliminary observations in the ACT model of IBD suggest only a transient benefit of oral TPX6001 treatment. However, several studies report activities of oral TGFβ, when given as an intact protein. Shiou et al[71] reported that oral administration of TGFβ (30 ng/mL) suppressed pro inflammatory cytokine production (including IL-6 and IL-8) in the gut of rat pups. The suppression was associated with suppressed NF-κB signaling. Systemic TGFβ levels were not measured. An earlier publication by Ando et al[72] reported increased serum TGFβ in mice after oral administration of the intact protein. Those studies also reported enhancement of oral tolerance. Additional studies in the ACT model, as well as other models of acute and chronic IBD, will be necessary to fairly evaluate the therapeutic potential of oral TPX6001 when given alone in IBD and perhaps also in other specific clinical situations where increasing TGFβ levels are pathogenic, for example against certain challenging forms of breast cancer[73]. Such studies are subjects of forthcoming work from our laboratories.

CONCLUSION

These observations suggest a negative feedback mechanism in the gut whereby local delivery of TGFβ results in reduced local and systemic levels of the active form of TGFβ. Our findings suggest potential clinical implications for use of encapsulated TGFβ, perhaps in the context of IBD and/or other instances of fibrosis and/or pathological TGFβ signaling.

ARTICLE HIGHLIGHTS
Research background

TreXTAM® is a combination of transforming growth factor beta (TGFβ) and all trans retinoic acid (ATRA) microencapsulated for oral delivery to immune structures of the gut. It is in development as a novel treatment for inflammatory bowel disease (IBD).

Research motivation

When given together, ATRA and TGFβ signals synergize in promoting the differentiation and stabilization of regulatory T cells.

Research objectives

This is a completely novel strategy for the treatment of IBD, as no similar products currently exist. TreXTAM would represent an entirely novel IBD treatment modality.

Research methods

During TreXTAM development, we studied TGFβ pharmacokinetics after oral administration of TreXTAM, or after the encapsulated cytokine (TPX6001) was given alone, without ATRA. This is required for combinatorial products.

Research results

We made the surprising discovery that oral administration of TreXTAM dramatically reduced TGFβ levels in colon and in blood, to below baseline levels. When encapsulated TGFβ (TPX6001) was given alone, three times a week for 25 d, we likewise observed serum TGFβ decreases below baseline (untreated) levels. Oral treatment with TPX6001 alone transiently ameliorated weight loss in the murine adoptive cell transfer model of IBD, chosen because it recapitulates regulatory T cell immunology associated with disease.

Research conclusions

These observations suggest a negative feedback mechanism in the gut whereby local delivery of TGFβ results in reduced local and systemic levels of the active form of TGFβ. This finding suggests potential clinical implications for use of encapsulated TGFβ in the context of IBD and/or pathological TGFβ signaling.

Research perspectives

Additional studies in the ACT model, as well as other models of acute and chronic IBD, will be necessary to fairly evaluate the therapeutic potential of oral TreXTAM, as well as TPX6001 when given alone in IBD, autoimmune diseases, and perhaps also in other specific clinical situations where increasing TGFβ levels are pathogenic, for example against certain challenging forms of breast cancer. Such studies are subjects of forthcoming work from our laboratories.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Sinagra E S-Editor: Gong ZM L-Editor: A P-Editor: Li JH

References
1.  Conway TF, Hammer L, Furtado S, Mathiowitz E, Nicoletti F, Mangano K, Egilmez NK, Auci DL. Oral Delivery of Particulate Transforming Growth Factor Beta 1 and All-Trans Retinoic Acid Reduces Gut Inflammation in Murine Models of Inflammatory Bowel Disease. J Crohns Colitis. 2015;9:647-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
2.  Auci DL, Egilmez NK. Synergy of Transforming Growth Factor Beta 1 and All Trans Retinoic Acid in the Treatment of Inflammatory Bowel Disease: Role of Regulatory T cells. J Gastroenterol Pancreatol Liver Disord. 2016;3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
3.  Yamanaka YJ, Leong KW. Engineering strategies to enhance nanoparticle-mediated oral delivery. J Biomater Sci Polym Ed. 2008;19:1549-1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 45]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
4.  Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature. 2009;458:1180-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 430]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
5.  Pertuit D, Moulari B, Betz T, Nadaradjane A, Neumann D, Ismaïli L, Refouvelet B, Pellequer Y, Lamprecht A. 5-amino salicylic acid bound nanoparticles for the therapy of inflammatory bowel disease. J Control Release. 2007;123:211-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 77]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
6.  Mathiowitz E, Jacob JS, Jong YS, Carino GP, Chickering DE, Chaturvedi P, Santos CA, Vijayaraghavan K, Montgomery S, Bassett M, Morrell C. Biologically erodable microspheres as potential oral drug delivery systems. Nature. 1997;386:410-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 668]  [Cited by in F6Publishing: 682]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
7.  Chung AY, Li Q, Blair SJ, De Jesus M, Dennis KL, LeVea C, Yao J, Sun Y, Conway TF, Virtuoso LP, Battaglia NG, Furtado S, Mathiowitz E, Mantis NJ, Khazaie K, Egilmez NK. Oral interleukin-10 alleviates polyposis via neutralization of pathogenic T-regulatory cells. Cancer Res. 2014;74:5377-5385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
8.  Benoit MA, Baras B, Gillard J. Preparation and characterization of protein-loaded poly(epsilon-caprolactone) microparticles for oral vaccine delivery. Int J Pharm. 1999;184:73-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 152]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
9.  Chu LY, Xie R, Zhu JH, Chen WM, Yamaguchi T, Nakao S. Study of SPG membrane emulsification processes for the preparation of monodisperse core-shell microcapsules. J Colloid Interface Sci. 2003;265:187-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 72]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
10.  Lee SC, Oh JT, Jang MH, Chung SI. Quantitative analysis of polyvinyl alcohol on the surface of poly(D, L-lactide-co-glycolide) microparticles prepared by solvent evaporation method: effect of particle size and PVA concentration. J Control Release. 1999;59:123-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 100]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
11.  Deng X, Zhou S, Li X, Zhao J, Yuan M. In vitro degradation and release profiles for poly-dl-lactide-poly(ethylene glycol) microspheres containing human serum albumin. J Control Release. 2001;71:165-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
12.  Ma GH, Su ZG, Omi S, Sundberg D, Stubbs J. Microencapsulation of oil with poly(styrene-N,N-dimethylaminoethyl methacrylate) by SPG emulsification technique: effects of conversion and composition of oil phase. J Colloid Interface Sci. 2003;266:282-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 62]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
13.  Supsakulchai A, Ma GH, Nagai M, Omi S. Preparation of uniform titanium dioxide (TiO2) polystyrene-based composite particles using the glass membrane emulsification process with a subsequent suspension polymerization. J Microencapsul. 2003;20:1-18.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Wei G, Pettway GJ, McCauley LK, Ma PX. The release profiles and bioactivity of parathyroid hormone from poly(lactic-co-glycolic acid) microspheres. Biomaterials. 2004;25:345-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 102]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
15.  Yang YY, Chung TS, Ng NP. Morphology, drug distribution, and in vitro release profiles of biodegradable polymeric microspheres containing protein fabricated by double-emulsion solvent extraction/evaporation method. Biomaterials. 2001;22:231-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 543]  [Cited by in F6Publishing: 490]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
16.  Freitas S, Merkle HP, Gander B. Ultrasonic atomisation into reduced pressure atmosphere--envisaging aseptic spray-drying for microencapsulation. J Control Release. 2004;95:185-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
17.  He P, Davis SS, Illum L. Chitosan microspheres prepared by spray drying. Int J Pharm. 1999;187:53-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in F6Publishing: 263]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
18.  Mu L, Feng SS. Fabrication, characterization and in vitro release of paclitaxel (Taxol) loaded poly (lactic-co-glycolic acid) microspheres prepared by spray drying technique with lipid/cholesterol emulsifiers. J Control Release. 2001;76:239-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 227]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
19.  Quaglia F, De Rosa G, Granata E, Ungaro F, Fattal E, Immacolata La Rotonda M. Feeding liquid, non-ionic surfactant and cyclodextrin affect the properties of insulin-loaded poly(lactide-co-glycolide) microspheres prepared by spray-drying. J Control Release. 2003;86:267-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 74]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
20.  Wang FJ, Wang CH. Sustained release of etanidazole from spray dried microspheres prepared by non-halogenated solvents. J Control Release. 2002;81:263-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 70]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
21.  Sharma A, Harper CM, Hammer L, Nair RE, Mathiowitz E, Egilmez NK. Characterization of cytokine-encapsulated controlled-release microsphere adjuvants. Cancer Biother Radiopharm. 2004;19:764-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
22.  Egilmez NK, Jong YS, Iwanuma Y, Jacob JS, Santos CA, Chen FA, Mathiowitz E, Bankert RB. Cytokine immunotherapy of cancer with controlled release biodegradable microspheres in a human tumor xenograft/SCID mouse model. Cancer Immunol Immunother. 1998;46:21-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
23.  Egilmez NK, Jong YS, Hess SD, Jacob JS, Mathiowitz E, Bankert RB. Cytokines delivered by biodegradable microspheres promote effective suppression of human tumors by human peripheral blood lymphocytes in the SCID-Winn model. J Immunother. 2000;23:190-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
24.  Egilmez NK, Jong YS, Sabel MS, Jacob JS, Mathiowitz E, Bankert RB. In situ tumor vaccination with interleukin-12-encapsulated biodegradable microspheres: induction of tumor regression and potent antitumor immunity. Cancer Res. 2000;60:3832-3837.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Hill HC, Conway TF Jr, Sabel MS, Jong YS, Mathiowitz E, Bankert RB, Egilmez NK. Cancer immunotherapy with interleukin 12 and granulocyte-macrophage colony-stimulating factor-encapsulated microspheres: coinduction of innate and adaptive antitumor immunity and cure of disseminated disease. Cancer Res. 2002;62:7254-7263.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Nair RE, Jong YS, Jones SA, Sharma A, Mathiowitz E, Egilmez NK. IL-12 + GM-CSF microsphere therapy induces eradication of advanced spontaneous tumors in her-2/neu transgenic mice but fails to achieve long-term cure due to the inability to maintain effector T-cell activity. J Immunother. 2006;29:10-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
27.  Florence AT. The oral absorption of micro- and nanoparticulates: neither exceptional nor unusual. Pharm Res. 1997;14:259-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 399]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
28.  Ermak TH, Dougherty EP, Bhagat HR, Kabok Z, Pappo J. Uptake and transport of copolymer biodegradable microspheres by rabbit Peyer's patch M cells. Cell Tissue Res. 1995;279:433-436.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 89]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
29.  Jani P, Halbert GW, Langridge J, Florence AT. The uptake and translocation of latex nanospheres and microspheres after oral administration to rats. J Pharm Pharmacol. 1989;41:809-812.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 263]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
30.  Egilmez NK, Jong YS, Mathiowitz E, Bankert RB. Tumor vaccination with cytokine-encapsulated microspheres. Methods Mol Med. 2003;75:687-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 12]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
31.  Jeong YI, Song JG, Kang SS, Ryu HH, Lee YH, Choi C, Shin BA, Kim KK, Ahn KY, Jung S. Preparation of poly(DL-lactide-co-glycolide) microspheres encapsulating all-trans retinoic acid. Int J Pharm. 2003;259:79-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 77]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
32.  Coombes JL, Maloy KJ. Control of intestinal homeostasis by regulatory T cells and dendritic cells. Semin Immunol. 2007;19:116-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
33.  Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ. All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J Exp Med. 2007;204:1765-1774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 625]  [Cited by in F6Publishing: 657]  [Article Influence: 38.6]  [Reference Citation Analysis (0)]
34.  Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775-1785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1408]  [Cited by in F6Publishing: 1437]  [Article Influence: 84.5]  [Reference Citation Analysis (0)]
35.  Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, Cheroutre H. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317:256-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1505]  [Cited by in F6Publishing: 1530]  [Article Influence: 90.0]  [Reference Citation Analysis (0)]
36.  Auci DL, Egilmez NK, Dryden GW. Anti-Fibrotic Potential of All Trans Retinoic Acid in Inflammatory Bowel Disease. J Gastroenterol Pancreatol Liver Disord. 2018;6 .  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
37.  Knabbe C, Klein H, Zugmaier G, Voigt KD. Hormonal regulation of transforming growth factor beta-2 expression in human prostate cancer. J Steroid Biochem Mol Biol. 1993;47:137-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
38.  Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009;361:2066-2078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1967]  [Cited by in F6Publishing: 2028]  [Article Influence: 135.2]  [Reference Citation Analysis (5)]
39.  Podolsky DK. Inflammatory bowel disease. N Engl J Med. 2002;347:417-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2693]  [Cited by in F6Publishing: 2685]  [Article Influence: 122.0]  [Reference Citation Analysis (2)]
40.  Kraus S, Arber N. Inflammation and colorectal cancer. Curr Opin Pharmacol. 2009;9:405-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 182]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
41.  Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nat Rev Immunol. 2008;8:458-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 666]  [Cited by in F6Publishing: 659]  [Article Influence: 41.2]  [Reference Citation Analysis (0)]
42.  Rutgeerts P, Vermeire S, Van Assche G. Biological therapies for inflammatory bowel diseases. Gastroenterology. 2009;136:1182-1197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 291]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
43.  Cannom RR, Kaiser AM, Ault GT, Beart RW Jr, Etzioni DA. Inflammatory bowel disease in the United States from 1998 to 2005: has infliximab affected surgical rates? Am Surg. 2009;75:976-980.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Rubtsov YP, Rudensky AY. TGFbeta signalling in control of T-cell-mediated self-reactivity. Nat Rev Immunol. 2007;7:443-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 246]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
45.  Li MO, Sanjabi S, Flavell RA. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity. 2006;25:455-471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 577]  [Cited by in F6Publishing: 621]  [Article Influence: 34.5]  [Reference Citation Analysis (0)]
46.  Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-beta receptor. Immunity. 2006;25:441-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 365]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
47.  He MX, He YW. Suppressing autoimmunity by TGF-β: not just through T(reg) cells. Cell Mol Immunol. 2012;9:371-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
48.  Chernajovsky Y, Adams G, Triantaphyllopoulos K, Ledda MF, Podhajcer OL. Pathogenic lymphoid cells engineered to express TGF beta 1 ameliorate disease in a collagen-induced arthritis model. Gene Ther. 1997;4:553-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 83]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
49.  Kuruvilla AP, Shah R, Hochwald GM, Liggitt HD, Palladino MA, Thorbecke GJ. Protective effect of transforming growth factor beta 1 on experimental autoimmune diseases in mice. Proc Natl Acad Sci USA. 1991;88:2918-2921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 295]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
50.  Moritani M, Yoshimoto K, Wong SF, Tanaka C, Yamaoka T, Sano T, Komagata Y, Miyazaki J, Kikutani H, Itakura M. Abrogation of autoimmune diabetes in nonobese diabetic mice and protection against effector lymphocytes by transgenic paracrine TGF-beta1. J Clin Invest. 1998;102:499-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 64]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
51.  Racke MK, Dhib-Jalbut S, Cannella B, Albert PS, Raine CS, McFarlin DE. Prevention and treatment of chronic relapsing experimental allergic encephalomyelitis by transforming growth factor-beta 1. J Immunol. 1991;146:3012-3017.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Zhang K, Phan SH. Cytokines and pulmonary fibrosis. Biol Signals. 1996;5:232-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 98]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
53.  Anscher MS, Kong FM, Jirtle RL. The relevance of transforming growth factor beta 1 in pulmonary injury after radiation therapy. Lung Cancer. 1998;19:109-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
54.  Matrat M, Lardot C, Huaux F, Broeckaert F, Lison D. Role of urokinase in the activation of macrophage-associated TGF-beta in silica-induced lung fibrosis. J Toxicol Environ Health A. 1998;55:359-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
55.  Zhang JG, Walmsley MW, Moy JV, Cunningham AC, Talbot D, Dark JH, Kirby JA. Differential effects of cyclosporin A and tacrolimus on the production of TGF-beta: implications for the development of obliterative bronchiolitis after lung transplantation. Transpl Int. 1998;11 Suppl 1:S325-S327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 7]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
56.  Haustein UF, Anderegg U. Pathophysiology of scleroderma: an update. J Eur Acad Dermatol Venereol. 1998;11:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Liem LM, Fibbe WE, van Houwelingen HC, Goulmy E. Serum transforming growth factor-beta1 levels in bone marrow transplant recipients correlate with blood cell counts and chronic graft-versus-host disease. Transplantation. 1999;67:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 60]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
58.  Kitamura M, Sütö TS. TGF-beta and glomerulonephritis: anti-inflammatory versus prosclerotic actions. Nephrol Dial Transplant. 1997;12:669-679.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 64]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
59.  Prud'homme GJ, Piccirillo CA. The inhibitory effects of transforming growth factor-beta-1 (TGF-beta1) in autoimmune diseases. J Autoimmun. 2000;14:23-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 214]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
60.  Song X, Liu W, Xie S, Wang M, Cao G, Mao C, Lv C. All-transretinoic acid ameliorates bleomycin-induced lung fibrosis by downregulating the TGF-β1/Smad3 signaling pathway in rats. Lab Invest. 2013;93:1219-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
61.  Frenz DA, Liu W. Treatment with all-trans-retinoic acid decreases levels of endogenous TGF-beta(1) in the mesenchyme of the developing mouse inner ear. Teratology. 2000;61:297-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
62.  Yu Z, Xing Y. All-trans retinoic acid inhibited chondrogenesis of mouse embryonic palate mesenchymal cells by down-regulation of TGF-beta/Smad signaling. Biochem Biophys Res Commun. 2006;340:929-934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
63.  Wang H, Dan Z, Jiang H. Effect of all-trans retinoic acid on liver fibrosis induced by common bile duct ligation in rats. J Huazhong Univ Sci Technolog Med Sci. 2008;28:553-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
64.  Tabata C, Tabata R, Hirayama N, Yasumitsu A, Yamada S, Murakami A, Iida S, Tamura K, Terada T, Kuribayashi K, Fukuoka K, Nakano T. All-trans-retinoic acid inhibits tumour growth of malignant pleural mesothelioma in mice. Eur Respir J. 2009;34:1159-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
65.  Evanson JR, Guyton MK, Oliver DL, Hire JM, Topolski RL, Zumbrun SD, McPherson JC, Bojescul JA. Gender and age differences in growth factor concentrations from platelet-rich plasma in adults. Mil Med. 2014;179:799-805.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 76]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
66.  Willis EL, Wolf RF, White GL, McFarlane D. Age- and gender-associated changes in the concentrations of serum TGF-1β, DHEA-S and IGF-1 in healthy captive baboons (Papio hamadryas anubis). Gen Comp Endocrinol. 2014;195:21-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
67.  Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol. 2017;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 333]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
68.  Wakefield LM, Winokur TS, Hollands RS, Christopherson K, Levinson AD, Sporn MB. Recombinant latent transforming growth factor beta 1 has a longer plasma half-life in rats than active transforming growth factor beta 1, and a different tissue distribution. J Clin Invest. 1990;86:1976-1984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 236]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
69.  Mazzieri R, Jurukovski V, Obata H, Sung J, Platt A, Annes E, Karaman-Jurukovska N, Gleizes PE, Rifkin DB. Expression of truncated latent TGF-beta-binding protein modulates TGF-beta signaling. J Cell Sci. 2005;118:2177-2187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
70.  Okuno M, Moriwaki H, Imai S, Muto Y, Kawada N, Suzuki Y, Kojima S. Retinoids exacerbate rat liver fibrosis by inducing the activation of latent TGF-beta in liver stellate cells. Hepatology. 1997;26:913-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 33]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
71.  Shiou SR, Yu Y, Guo Y, Westerhoff M, Lu L, Petrof EO, Sun J, Claud EC. Oral administration of transforming growth factor-β1 (TGF-β1) protects the immature gut from injury via Smad protein-dependent suppression of epithelial nuclear factor κB (NF-κB) signaling and proinflammatory cytokine production. J Biol Chem. 2013;288:34757-34766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
72.  Ando T, Hatsushika K, Wako M, Ohba T, Koyama K, Ohnuma Y, Katoh R, Ogawa H, Okumura K, Luo J, Wyss-Coray T, Nakao A. Orally administered TGF-beta is biologically active in the intestinal mucosa and enhances oral tolerance. J Allergy Clin Immunol. 2007;120:916-923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
73.  Bhola NE, Balko JM, Dugger TC, Kuba MG, Sánchez V, Sanders M, Stanford J, Cook RS, Arteaga CL. TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest. 2013;123:1348-1358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 415]  [Cited by in F6Publishing: 446]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]