Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
15th National Conference of the IAOMFP, Chennai, 2006
Abstract
Abstracts from current literature
Acne in India: Guidelines for management - IAA Consensus Document
Addendum
Announcement
Art & Psychiatry
Article
Articles
Association Activities
Association Notes
Award Article
Book Review
Brief Report
Case Analysis
Case Letter
Case Letters
Case Notes
Case Report
Case Reports
Clinical and Laboratory Investigations
Clinical Article
Clinical Studies
Clinical Study
Commentary
Conference Oration
Conference Summary
Continuing Medical Education
Correspondence
Corrigendum
Cosmetic Dermatology
Cosmetology
Current Best Evidence
Current Issue
Current View
Derma Quest
Dermato Surgery
Dermatopathology
Dermatosurgery Specials
Dispensing Pearl
Do you know?
Drug Dialogues
e-IJDVL
Editor Speaks
Editorial
Editorial Remarks
Editorial Report
Editorial Report - 2007
Editorial report for 2004-2005
Errata
Erratum
Focus
Fourth All India Conference Programme
From Our Book Shelf
From the Desk of Chief Editor
General
Get Set for Net
Get set for the net
Guest Article
Guest Editorial
History
How I Manage?
IADVL Announcement
IADVL Announcements
IJDVL Awards
IJDVL AWARDS 2015
IJDVL Awards 2018
IJDVL Awards 2019
IJDVL Awards 2020
IJDVL International Awards 2018
Images in Clinical Practice
In Memorium
Inaugural Address
Index
Knowledge From World Contemporaries
Leprosy Section
Letter in Response to Previous Publication
Letter to Editor
Letter to the Editor
Letter to the Editor - Case Letter
Letter to the Editor - Letter in Response to Published Article
LETTER TO THE EDITOR - LETTERS IN RESPONSE TO PUBLISHED ARTICLES
Letter to the Editor - Observation Letter
Letter to the Editor - Study Letter
Letter to the Editor - Therapy Letter
Letter to the Editor: Articles in Response to Previously Published Articles
Letters in Response to Previous Publication
Letters to the Editor
Letters to the Editor - Letter in Response to Previously Published Articles
Letters to the Editor: Case Letters
Letters to the Editor: Letters in Response to Previously Published Articles
Medicolegal Window
Messages
Miscellaneous Letter
Musings
Net Case
Net case report
Net Image
Net Images
Net Letter
Net Quiz
Net Study
New Preparations
News
News & Views
Obituary
Observation Letter
Observation Letters
Oration
Original Article
ORIGINAL CONTRIBUTION
Original Contributions
Pattern of Skin Diseases
Pearls
Pediatric Dermatology
Pediatric Rounds
Perspective
Presedential Address
Presidential Address
Presidents Remarks
Quiz
Recommendations
Regret
Report
Report of chief editor
Report of Hon : Treasurer IADVL
Report of Hon. General Secretary IADVL
Research Methdology
Research Methodology
Resident page
Resident's Page
Resident’s Page
Residents' Corner
Residents' Corner
Residents' Page
Retraction
Review
Review Article
Review Articles
Reviewers 2022
Revision Corner
Self Assessment Programme
SEMINAR
Seminar: Chronic Arsenicosis in India
Seminar: HIV Infection
Short Communication
Short Communications
Short Report
Snippets
Special Article
Specialty Interface
Studies
Study Letter
Study Letters
Supplement-Photoprotection
Supplement-Psoriasis
Symposium - Contact Dermatitis
Symposium - Lasers
Symposium - Pediatric Dermatoses
Symposium - Psoriasis
Symposium - Vesicobullous Disorders
SYMPOSIUM - VITILIGO
Symposium Aesthetic Surgery
Symposium Dermatopathology
Symposium-Hair Disorders
Symposium-Nails Part I
Symposium-Nails-Part II
Systematic Review and Meta-Analysis
Systematic Reviews and Meta-analyses
Systematic Reviews and Meta-analysis
Tables
Technology
Therapeutic Guideline-IADVL
Therapeutic Guidelines
Therapeutic Guidelines - IADVL
Therapeutics
Therapy
Therapy Letter
Therapy Letters
View Point
Viewpoint
What’s new in Dermatology
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
15th National Conference of the IAOMFP, Chennai, 2006
Abstract
Abstracts from current literature
Acne in India: Guidelines for management - IAA Consensus Document
Addendum
Announcement
Art & Psychiatry
Article
Articles
Association Activities
Association Notes
Award Article
Book Review
Brief Report
Case Analysis
Case Letter
Case Letters
Case Notes
Case Report
Case Reports
Clinical and Laboratory Investigations
Clinical Article
Clinical Studies
Clinical Study
Commentary
Conference Oration
Conference Summary
Continuing Medical Education
Correspondence
Corrigendum
Cosmetic Dermatology
Cosmetology
Current Best Evidence
Current Issue
Current View
Derma Quest
Dermato Surgery
Dermatopathology
Dermatosurgery Specials
Dispensing Pearl
Do you know?
Drug Dialogues
e-IJDVL
Editor Speaks
Editorial
Editorial Remarks
Editorial Report
Editorial Report - 2007
Editorial report for 2004-2005
Errata
Erratum
Focus
Fourth All India Conference Programme
From Our Book Shelf
From the Desk of Chief Editor
General
Get Set for Net
Get set for the net
Guest Article
Guest Editorial
History
How I Manage?
IADVL Announcement
IADVL Announcements
IJDVL Awards
IJDVL AWARDS 2015
IJDVL Awards 2018
IJDVL Awards 2019
IJDVL Awards 2020
IJDVL International Awards 2018
Images in Clinical Practice
In Memorium
Inaugural Address
Index
Knowledge From World Contemporaries
Leprosy Section
Letter in Response to Previous Publication
Letter to Editor
Letter to the Editor
Letter to the Editor - Case Letter
Letter to the Editor - Letter in Response to Published Article
LETTER TO THE EDITOR - LETTERS IN RESPONSE TO PUBLISHED ARTICLES
Letter to the Editor - Observation Letter
Letter to the Editor - Study Letter
Letter to the Editor - Therapy Letter
Letter to the Editor: Articles in Response to Previously Published Articles
Letters in Response to Previous Publication
Letters to the Editor
Letters to the Editor - Letter in Response to Previously Published Articles
Letters to the Editor: Case Letters
Letters to the Editor: Letters in Response to Previously Published Articles
Medicolegal Window
Messages
Miscellaneous Letter
Musings
Net Case
Net case report
Net Image
Net Images
Net Letter
Net Quiz
Net Study
New Preparations
News
News & Views
Obituary
Observation Letter
Observation Letters
Oration
Original Article
ORIGINAL CONTRIBUTION
Original Contributions
Pattern of Skin Diseases
Pearls
Pediatric Dermatology
Pediatric Rounds
Perspective
Presedential Address
Presidential Address
Presidents Remarks
Quiz
Recommendations
Regret
Report
Report of chief editor
Report of Hon : Treasurer IADVL
Report of Hon. General Secretary IADVL
Research Methdology
Research Methodology
Resident page
Resident's Page
Resident’s Page
Residents' Corner
Residents' Corner
Residents' Page
Retraction
Review
Review Article
Review Articles
Reviewers 2022
Revision Corner
Self Assessment Programme
SEMINAR
Seminar: Chronic Arsenicosis in India
Seminar: HIV Infection
Short Communication
Short Communications
Short Report
Snippets
Special Article
Specialty Interface
Studies
Study Letter
Study Letters
Supplement-Photoprotection
Supplement-Psoriasis
Symposium - Contact Dermatitis
Symposium - Lasers
Symposium - Pediatric Dermatoses
Symposium - Psoriasis
Symposium - Vesicobullous Disorders
SYMPOSIUM - VITILIGO
Symposium Aesthetic Surgery
Symposium Dermatopathology
Symposium-Hair Disorders
Symposium-Nails Part I
Symposium-Nails-Part II
Systematic Review and Meta-Analysis
Systematic Reviews and Meta-analyses
Systematic Reviews and Meta-analysis
Tables
Technology
Therapeutic Guideline-IADVL
Therapeutic Guidelines
Therapeutic Guidelines - IADVL
Therapeutics
Therapy
Therapy Letter
Therapy Letters
View Point
Viewpoint
What’s new in Dermatology
View/Download PDF

Translate this page into:

Review Article
87 (
6
); 753-767
doi:
10.25259/IJDVL_19_20
pmid:
34245532

Stem cell therapy in dermatology

Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India
Corresponding author: Dr. Sujay Khandpur, Department of Dermatology and Venereology, All India Institute of Medical Sciences, New Delhi, India. sujay_khandpur@yahoo.com
Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Khandpur S, Gupta S, Gunaabalaji DR. Stem cell therapy in dermatology. Indian J Dermatol Venereol Leprol 2021;87:753-67.

Abstract

Stem cells are precursor cells present in many tissues with ability to differentiate into various types of cells. This interesting property of plasticity can have therapeutic implications and there has been substantial research in this field in last few decades. As a result, stem cell therapy is now used as a therapeutic modality in many conditions, and has made its way in dermatology too. Stem cells can be classified on the basis of their source and differentiating capacity. In skin, they are present in the inter-follicular epidermis, hair follicle, dermis and adipose tissue, which help in maintaining normal skin homeostasis and repair and regeneration during injury. In view of their unique properties, they have been employed in treatment of several dermatoses including systemic sclerosis, systemic lupus erythematosus, scleromyxedema, alopecia, Merkel cell carcinoma, pemphigus vulgaris, psoriasis, wound healing, epidermolysis bullosa and even aesthetic medicine, with variable success. The advent of stem cell therapy has undoubtedly brought us closer to curative treatment of disorders previously considered untreatable. Nevertheless, there are multiple lacunae which need to be addressed including ideal patient selection, timing of intervention, appropriate conditioning regimens, post-intervention care and cost effectiveness. Further research in these aspects would help optimize the results of stem cell therapy.

Keywords

Dermatology
pemphigus
stem cell therapy
systemic lupus erythematosus
systemic sclerosis

Introduction

Stem cell therapy is a novel technique which had gained significant attention over the past years. The Nobel Prize in Physiology or Medicine 2007 was awarded jointly to Mario R. Capecchi, Sir Martin J. Evans and Oliver Smithies for their discoveries of principles for introducing specific gene modifications in mice by the use of embryonic stem cells.1 Stem cells have been employed in broad therapeutic indications and as a consequence, the New Drugs and Clinical Trials Rules include “stem cell derived products” under the new drug section.2 In dermatology, stem cell therapy has been tried in several refractory conditions with some success which has widened the therapeutic armamentarium. In this article, we aim to review the current status of stem cell therapy in dermatology.

Materials and Methods

A search for relevant literature in English language was conducted using PubMed, MEDLINE, Hindawi and Google Scholar. All publications up to 2019 were identified using the following key words: stem cell therapy, dermatology, hematopoietic, autologous and allogenic. All articles, including case reports, case series, randomized controlled trials and review articles on the use of stem cell therapy in various dermatological conditions, were considered and the results of the studies including the adverse effects were tabulated. We included articles related to clinical relevance of stem cells and stem cell therapy in dermatology. We excluded articles that were outside the domain of clinical dermatology, such as those on basic science of stem cells, veterinary dermatology and use of stem cells in other fields of medicine. The guidelines by European Group for Blood and Marrow Transplantation (EBMT), British Society of Blood and Marrow Transplantation (BSBMT), American Society for Blood and Marrow Transplantation (ASBMT) and Indian Council of Medical Research (ICMR) were compiled to give a comprehensive overview of indications and current status of stem cell therapy in dermatology. We were able to review about 110 articles on this subject which form the basis of this review article.

Principles of Stem Cell Therapy

Stem cells are undifferentiated cells present in different organ systems with the three hallmark characteristics of self-renewal, differentiation and plasticity [Table 1].3 Stem cells are present in a specialized microenvironment called stem cell niche which functions as a unit to maintain homeostasis and repair tissues at the time of injury.4 Stem cells can be classified on the basis of their source and differentiating capacity [Table 2].5-7

Table 1: Defining characteristics of a stem cell3
Property Definition
Self-renewal Ability to undergo numerous cycles of asymmetrical cell division to produce differentiated cells as well as cells that are similar to the parent cell, thereby maintaining the pool of undifferentiated cells
Differentiation Ability to differentiate into cells of the tissue in which stem cell is located
Plasticity/trans-differentiation Ability of adult stem cells to cross lineage barriers and differentiate into cells of tissue different from the original tissue
Table 2: Classification of stem cells5-7
Classification Description
Classification on the basis of differentiating capacity Totipotent/omnipotent The ability to differentiate into all possible cell types including placenta
Cells produced by the first few divisions of the fertilized egg, known as morula cells, are totipotent
Pluripotent The ability of a stem cell to turn into all mature cell types of the body of all the three germ layers, except placenta
Embryonic stem cells that are isolated from an early stage embryo, called blastocyst are pluripotent cells
Multipotent The ability to turn into more than one mature cell type of the body, usually a restricted and related group of different cell types
E.g., hematopoietic (adult) stem cells that can become red and white blood cells or platelets
Mesenchymal stem cells that can become a wide variety, but related group, of mature cell types (bone, cartilage, connective tissue, and adipose tissue)
Oligopotent The ability to differentiate into a few cells.
Example: Adult lymphoid or myeloid stem cells.
Unipotent Can produce only one cell type
Muscle stem cells
Classification on the basis of their source Embryonic stem cell Pluripotent stem cells present in the inner cell mass of blastocyst which are capable of producing all organs in human body except for placenta
Somatic/adult stem cell Somatic stem cells (multipotent or unipotent), with limited plasticity, present in many tissues including skin and bone marrow
iPSC Recently, the induced pleuripotent stem cells have emerged as a distinct variety. They are produced by reprogramming of somatic cells into pleuripotent state by genetic engineering. These stem cells can be induced to form various types of differentiated cells

iPSC: Induced pleuripotent stem cells

Epidermal Stem Cells

In skin, stem cells are found in inter-follicular epidermis (keratinocyte), bulge of the hair follicle (keratinocyte, melanocyte and neuronal), sebaceous gland, dermal papillae, dermis and subcutaneous tissue (mesenchymal). Melanocyte stem cells can be induced by ultraviolet radiation, laser, dermabrasion and drugs including tacrolimus, hence are used in re-pigmentation of vitiligo.8-13 Under normal circumstances, the stem cells from epidermis, hair follicle and sebaceous glands differentiate independent of each other. During injury, by virtue of plasticity, any stem cell from one location can give rise to whole cell lineage.14

Bone Marrow Stem Cells

Bone marrow contains hematopoietic and mesenchymal stem cells. The former can be derived from bone marrow and umbilical cord blood and are used in autoimmune disorders after immune ablation, deleting the self-reactive cells and repopulating with cells with improved self-tolerance. The hematopoietic stem cells can be derived from the same donor (autologous – as the loss of self-tolerance is at the peripheral blood level, but not the stem cell level as they are not involved in antigen recognition) or from an HLA matched donor (allogenic). Umbilical cord blood transplantation has few advantages over bone marrow transplant including lower incidence and lower severity of acute and chronic graft-versus-host disease, lower risk of transmitting latent virus infections and elimination of clinical risk to the donor during hematopoietic stem cell procurement procedures. However, disadvantages include higher risk of graft rejection and delayed hematopoietic recovery after transplantation due to a reduced number of hematopoietic progenitor cells that can further contribute to serious infections.15

Mesenchymal stem cells are multipotent adult stem cells derived from almost all tissues including bone marrow, umbilical cord, peripheral blood, skin, foreskin, fallopian tube, lung, fetal tissue, placenta, adipose tissue or amniotic fluid. In contrast to hematopietic stem cells, mesenchymal stem cells have low immunogenicity by virtue of their low MHC expression and also possess immunomodulatory effects, hence have utility in many diseases including inflammatory diseases. They also provide greater advantages over other stem cells including their relatively easy tissue isolation, absence of obvious risk for the donor or ethical constraints, capacity of migrating and homing to the injured site (e.g., tumor tropism), ability to expand for a relatively long period of time, ability to modify the host immune environment and higher transdifferentiation potential.16

The steps of allogenic hematopoietic stem cell therapy include stem cell mobilization, collection of stem cells, conditioning of recipient, stem cell infusion and recovery [Table 3].17-20

Table 3: Steps and principles of allogenic hematopoietic stem cell therapy17-20
Step Comments
Stem cell mobilization Quiescent hematopoietic stem cells in bone marrow are tethered to osteoblasts, other stromal cells and the extracellular matrix in the stem cell niche through a variety of adhesive molecule interactions. Stem cell mobilization aims at disruption of these niche interactions, thereby resulting in release of stem cell from the bone marrow into the peripheral blood. Mobilization regimens include cytotoxic agents (cyclophosphamide), hematopoietic growth factors (G-CSF, GM-CSF, both are FDA approved), small-molecule chemokine analogues (Plerixafor, an inhibitor of CXCR4, FDA approved for patients who fail to mobilize sufficient CD34+ cells for ASCT), recombinant monoclonal antibody (Natalizumab)
Collection of stem cells or harvesting The donor stem cell can be obtained by either direct bone marrow biopsy or by peripheral stem cell mobilization and collection of peripheral blood. Subsequently, either simple apheresis or stem cell manipulation with selection for CD34+ is done so as to prevent reinfusion of autoreactive cells. In autologous transplant the stem cells are stored in deep-freeze condition/in liquid nitrogen as a gap of 4 weeks is given for the cells to recover to undergo conditioning regimen, while in allogeneic transplant, collection and transplantation are done on the same day, eliminating the need for storage
Conditioning of recipient Preparative/conditioning regimen has to be given in autologous and allogeneic transplantation to prevent graft rejection and reduce tumor burden. The agents used include both radiotherapy and chemotherapy. Total myeloablation was the norm in older days, but with recognition of graft-versus-tumor effect having a contributory role in the success of allogenic HCT, reduced intensity, nonmyeloablative conditioning regimens have been developed nowadays for better acceptance. Examples of conditioning regime include cyclophosphamide, busulfan, fludarabine, antithymocyte globulin, cytosine arabinoside, anti-CD45 antibody conjugated to I-131, high-dose TBI (800–1320 cGy). Low-dose TBI (200–400 cGy)
Stem cell infusion Following conditioning, stem cells are thawed and reinfused, either into the bone marrow or through intravenous route. Even when given in peripheral circulation, the stem cells home to bone marrow and engraftment takes place
Recovery Following the conditioning regimen, the patient enters a stage of intense immunosuppression/myelosuppression, which is related to increased mortality and morbidity in transplant recipient. Supportive therapy is given which includes anti-emetics, anti-diarrheal, antibiotic, antiviral, and antifungal coverage. G-CSF, red cell and platelet transfusions is done when the levels of leukocytes are <1000, hemoglobin <7 g, platelet <30,000 respectively

ASCT: Autologous stem cell transplantation, CXCR4: Chemokine receptor type 4, CD34: Cluster of differentiation 4, cGy: Centigray, FDA: Food and drug administration, G-CSF: Granulocyte-colony stimulating factor, GM-CSF: Granulocyte-macrophage colony stimulating factor, HCT: Hematopoietic cell transplantation, TBI: Total body irradiation

Applications of Stem Cells in Dermatology[Table 4]

Pemphigus

Table 4: Dermatological conditions in which stem cell therapy has been tried
Good results Promising results Results with low evidence Discouraging results
Systemic sclerosis
SLE
Pemphigus
Psoriasis
Vitiligo
Wound healing
Scleromyxedema
Alopecia
HIV
Melanoma
Aesthetic medicine
Epidermolysis bullosa
Merkel cell carcinoma

SLE: Systemic lupus erythematosus

Even though the first-line treatment of pemphigus remains to be corticosteroids and other immunosuppressants, some patients remain refractory to therapy, making it imperative to explore other therapies. Hematopoietic stem cell therapy has been tried successfully in pemphigus as shown in a few studies.21-24 The proposed mechanism of action is that the transplanted stem cells repopulate the immune system, the number of autoreactive immune cells decline and this helps to restore the immunological balance.

There are case reports and small case series from India and abroad on successful treatment of recalcitrant pemphigus with both autologous and allogeneic hematopoietic stem cell therapy using different mobilization and conditioning regimens [Table 5]. Infection was the most common side effect, with sepsis and occasionally death occurring in these patients.21-24

Table 5: Studies on stem cell therapy in treatment of pemphigus
Author, year Indication and inclusion Methods Results Side effects
Oyama et al., 200421
Case report
Refractory pemphigus foliaceus. BSA - 20%
Resistant to topical betamethasone, oral prednisone, azathioprine, MMF, dapsone, cyclophosphamide 75 mg/day, for 9 months
Autologous HSCT
Mobilization - cyclophosphamide and G-CSF
Conditioning regimen: Cyclophosphamide and rATG. Methylprednisolone 1.0 g/day before each dose of rATG
Skin lesions resolved over 2 months; maintained till 10 months
Prednisone tapered off over 4 months
Culture negative neutropenic fever, nausea, anorexia
Relapse: Few erythematous plaques on nose and scalp, responded to topical steroids
No systemic therapy for 19 months post-HSCT
Suslova et al., 201022
Case report
Pemphigus vulgaris.
BSA - 30%
Resistant to steroids, methotrexate, dapsone, chlorambucil, azathioprine
Allogeneic HSCT
Conditioning regimen: Alemtuzumab, 300 cGy of TBI
Adjuvant: oral sirolimus
Severity decreased by 9thmonth post-transplantation Arthralgia arthritis
Relapse: None at 24 months
Vanikar et al., 201223
Case series, 11 patients
Clinical and biopsy proven pemphigus vulgaris, resistant to prednisolone and topical steroid Cytokine-stimulated allogeneic HSCT Recovery began (skin lesions started regressing) within 24h of HSCT and new lesions stopped erupting after 6 months. Over a mean follow-up of 8.02 years, all patients were well without recurrence/new lesions No GVHD/AE observed in any patient/donor
Wang et al., 201724
Case series, 12 patients
Nine of pemphigus vulgaris, three of pemphigus erythematosus, one of pemphigus foliaceus
Persisting disease after high doses of steroids, or at least one kind of immunosuppressant, for <6 months; or patients with steroid-related diseases or severe contraindications and complications to steroids
Autologous peripheral HSCT Overall survival was 91.6% at 80.33 months, complete remission was achieved and maintained in 90.9% (10/11); 81.8% (9/11) and, 75% (6/8) patients at 6 months, 1 year and 5 years, respectively,
two patients developed relapse at 5 and 6 months post-transplant
Infection (most common side effect) in 8 (66.7%) cases, with sepsis in 2 (16.7%), of which one died at 2 months post-transplant. Other AE: Pyrexia, headache and transaminitis

BSA: Body surface area, cGy: Centigray, G-CSF: Granulocyte-colony stimulating factor, GVHD: Graft versus host disease, HSCT: Hematopoietic stem cell transplantation, MMF: Mycophenolate mofetil, rATG: Rabbit antithymocyte globulin, TBI: Total body irradiation, AE: Adverse event

The available literature suggests the efficacy of hematopietic stem cell therapy in treatment of pemphigus (grade of recommendation C, level of evidence 4); however, large scale multicentric studies with longer follow-up are needed to confirm the results.

Systemic sclerosis

In search of a definite disease modifying treatment, systemic sclerosis is one of the first autoimmune diseases to be subjected to stem cell therapy. Hematopoietic stem cell therapy aims to non-specifically immunoablate aberrant self-reactive Tand B-cells through high-dose immunosuppression, with subsequent reconstitution of a renewed and tolerant immune system by means of infusing patient’s previously collected hematopoietic stem cells. Autologous hematopoietic stem cell therapy is preferred over allogeneic therapy due the lower treatment-related mortality and lack of graft-vs-host disease in the former.25 Stem cell therapy in this condition has been well studied in three randomized controlled trials (RCTs), namely, American Scleroderma Stem cell versus Immune Suppression Trial (ASSIST, phase 2, 19 patients), Autologous Stem cell Transplantation International Scleroderma Trial (ASTIS, phase 3, 156 patients) and The Scleroderma Cyclophosphamide Or Transplantation study (SCOT, phase 3, 75 patients), besides several case series and pilot studies.26-38 Despite the slight differences in methodology, all the studies have shown autologous hematopoietic stem cell therapy as an effective, safe and feasible modality in systemic sclerosis (level of evidence-2a, 2b. 4) [Table 6]. Since severe major organ involvement (pulmonary, cardiac or renal) or serious comorbidities are an absolute contraindication for hematopoietic stem cell therapy, these patients were excluded from all the three trials. It has been found to be more effective than conventional immunosuppressive therapies and is currently the only disease modifying strategy for improving long-term survival, prevention of organ worsening and improvement of skin and pulmonary function and improving the overall quality of life of patients.39 Data provided by the European and American registries include overall 3-year survival rates of around 80% and 5-year progression-free survival rate of 55%.29,40 The European Society for blood and marrow transplantation and British society of blood and marrow transplantation categorize autologous hematopoietic stem cell therapy in severe resistant disease as “clinical opinion,” that is, after assessing risks and benefits.41 Guidelines from the American society for blood and marrow transplantation categorize autologous hematopoietic stem cell therapy as “standard of care, rare indication” (as a treatment option for individual patients after careful evaluation of risks and benefits) in children and “developmental” in adults.42

Table 6: Studies on stem cell therapy in systemic sclerosis
Author, year Indication and inclusion Methods Results Side effects
(number of patients)
Oyama et al., 200726
Case series,
10 patients
Diffuse systemic sclerosis with an mRSS >13 and internal organ involvement Autologous HSCT
Mobilization: cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide plus rabbit anti-thymocyte globulin
Outcome measures: mRSS, PFT, HRCT, cardiac and renal
Significant improvement in mRSS at 6, 12 and 24 months
Cardiac, pulmonary and renal functions - Stable
Neutropenic fever - 5
Clostridium difficile colitis, culture negative pulmonary infiltrate, fluid overload, acute renal failure, engrafment syndrome - 1 patient each
chemotherapy-related nausea, vomiting, diarrhea, asthenia and mild liver enzyme elevation - Most of the patients
Nash et al., 200727
Case series,
34 patients
Early (<4 years) diffuse scleroderma (mRSS >15) and internal organ involvement Autologous HSCT
Mobilization: G-CSF
Conditioning regimen: Total body irradiation with cyclophosphamide and equine anti-thymocyte globulin
Post-transplant - prednisone
One-year survival - 79%
Significant improvement in mRSS and FVC
CMV gastroenteritis - 1
Bacteremia - 11
Herpes zoster - 6
Fatal pulmonary toxicities - 2
Renal crisis - 6
Supraventricular arrhythmias - 2 patients
Heart failure - 2
Treatment related death - 8 (23%) cases
Vonk et al., 200828
Retrospective study,
26 patients
Rapidly progressive disease (2 years duration), mRSS >20 or a disease duration >2 years, progression of mRSS (>20%) plus major organ involvement Autologous HSCT
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide plus anti-thymocyte globulin
Outcome measures: mRSS, PFT, survival
mRSS - significant decrease in mRSS in 19 (73%) cases after 1 year and in 15/16 (94%) patients after 5 years
No significant change in FEV1or DLCO
Progression/event free survival 64.3% at 5 years and 57.1% at 7 years
Transplant related mortality: 1 (3.8%) case
Relapse: 6 (28%) patients at 2–4 years
Farge et al., 201029
Retrospective study,
175 patients
Systemic sclerosis Autologous HSCT
Myeloablative (total body irradiation)/non-myeloablative (cyclophosphamide/busulfan/carmustine, cytarabine, melphalan, and etoposide/antithymocyte globulin)
Outcome measures: survival
Overall survival: 72.6% at 5 years
Progression/event free survival: 55% at 5 years
Transplant related mortality: 12 (6.8%) cases
Burt et al., 201130
Phase 2 ASSIST trial. 19 patients
Single center randomized controlled trial
Diffuse systemic sclerosis mRSS ≥15 with internal organ involvement, disease duration ≤4 years, age <60 years Autologous peripheral blood HSCT (ten patients) versus cyclophosphamide pulse (nine patients)
HSCT group: mobilization with cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide plus anti-thymocyte globulin
Control group: Cyclophosphamide 1 g/sq.m monthly pulse for 6 months. Outcome measures: improvement in mRSS (>25%) and FVC (>10%)
Mean follow up period of 2.6 years
All patients of stem cell group improved at or before 12 months follow-up. No patient had disease progression. Improvement in FVC and mRSS persisted at follow-up. In control group, none had improvement. Seven patients switched to receive stem cell transplantation, after which 4 were followed up for at least 1 year, with all showing improvement. No mortality in both groups at 12 months and overall Arrythmias (2 patients), volume overload (two patients), reactivation of CMV infection (1 patient)
Henes et al., 201231
Case series,
26 patients
Systemic sclerosis with inefficacy of cyclophosphamide or rapidly progressive diffuse disease Autologous stem cell transplantation
Mobilization: cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide plus antithymocyte globulin
Outcome measures: mRSS, PFT
Response rate 25% improvement in mRSS score in 21/23 (91%) patients 3 deaths before transplantation. Transplant-related mortality: 4%
Treatment-related mortality: 11%
Relapse: 7 patients (4.4 years of follow-up)
Burt et al., 201332
Retrospective study, 90 patients
Diffuse systemic sclerosis (mRSS >14) and internal organ involvement (pulmonary, cardiac or gastrointestinal) Autologous HSCT
Mobilization: cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide and rATG
Outcome measures: mRSS and PFT
mRSS and FVC improved. Total lung capacity and DLCO not improved Treatment related mortality: 5 (6%) cases
van Laar et al., 201433
ASTIS trial,
156 patients
Multicenter randomized controlled trial, Phase 3
Diffuse systemic sclerosis, mRSS ≥15 and internal organ involvement, disease duration ≤4 years, age 18–65 years Autologous HSCT (79 patients) versus cyclophosphamide pulses (77 patients). Mobilization: cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide plus anti-thymocyte globulin
Control group: cyclophosphamide 750 mg/sq.m monthly pulse for
12 months
Outcome measures: event free survival till death/persistent major organ failure
Median follow up 5.8 years
HRCT group - significant improvement in MRSS, total lung capacity, FVC
HSCT was associated with increased treatment-related mortality (8 cases, 10%), none in control group in the first year after treatment. However, HCST conferred significant long-term event-free survival benefit
Grade 3 or 4 AEs in first 2 years of follow-up: 51 (62.9%) patients in HSCT group and 30 (37%) in control group
Henes et al., 201434
Case series,
6 patients
Progressive systemic sclerosis with cardiac manifestations (biopsy proven myocardial fibrosis) Autologous HSCT
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: thiotepa, cyclophosphamide and rATG
Outcome measures: mRSS, HRCT chest and PFT
Significant improvement in mRSS score at 6 and 12 months. In 4 patients - >25% improvement
Median lung density and total lung volume improved
Non-significant improvement of FVC
Aspergillus pneumonia - 2 patients
No transplant related mortality
Relapse: 2 patients
Del Papa et al., 201735
Retrospective study, 18 patients
Rapidly progressing diffuse systemic sclerosis with disease duration <4 years
Control group (36 patients) treated with conventional therapies
Autologous HSCT
Mobilization: cyclophosphamide and filgrastim
Conditioning regimen: Cyclophosphamide and rATG
Outcome measures: mRSS, DLCO, disease activity using the ESSG scoring system
follow-up of 60 months
Statistically significant reduction in mRSS and DLCO in autologous HSCT group compared to control group at 1 year and maintained at the end of follow-up. Significantly lower mortality in autologous HSCT group (5.8%) compared to control (61%) Only one patient died during follow-up due to systemic sclerosis related manifestation (fatal cardiac arrhythmia occurring after 34 months)
Sullivan et al., 201836
SCOT trial, 75 patients
Multicenter randomized, open-label, phase 3 trial
Diffuse systemic sclerosis, mRSS ≥16 and internal organ involvement, disease duration ≤4 years, age 18–69 years Autologous HSCT (36 patients) versus cyclophosphamide (39 patients)
Mobilization: G-CSF
Conditioning regimen: Fractionated total-body irradiation, cyclophosphamide and equine antithymocyte globulin
Control group: cyclophosphamide 500 mg/sq.m followed by 750 mg/sq.m monthly pulse for 11 months
Outcome measures: Global rank composite score at 54 months
Follow up to 4.5 years
Rate of event-free survival at 54 months was 79% in transplantation group and 50% in cyclophosphamide group (P=0.02). At 72 months, Kaplan–Meier estimates of event-free survival (74% vs. 47%) and overall survival (86% vs. 51%) also favored transplantation (P=0.03 and 0.02, respectively). Treatment-related mortality in the transplantation group was 3% at 54 months and 6% at 72 months, as compared with none in cyclophosphamide group Rate of serious AEs in person-years: 0.38 (transplantation group) and 0.52 (cyclophosphamide group). Rate of infections (of any grade) per person-year: 0.75 (transplantation group) and 0.79 (cyclophosphamide group)
Rate of infections of grade 3 or more per person-year: 0.21 (transplantation group) and 0.13 (cyclophosphamide group)
Nakamura et al., 201837
14 patients
Long-term follow-up in a phase II Trial
SSc patients with disease duration <3 years, with at least one of the following: mRSS) ≥15, refractory digital ulcer or interstitial lung disease HSCT were performed after conditioning using cyclophosphamide
Median follow-up period was 137 months
Overall survival 93%, event-free survival rate 40% at 10 years. Eight patients (57%) achieved more than 50% decrease in mRSS from baseline within 6 months after HSCT AEs related to HSCT occurred in 6 patients (43%). Severe cardiomyopathy occurred in 2 patients, and one of them had a fatal course
Nair et al., 201838
Case series,
4 patients
Diffuse systemic sclerosis with mRSS of 15 and one internal organ involvement Autologous HSCT
Mobilization: cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide plus fludarabine plus anti-thymocyte globulin
Outcome measures: mRSS, PFT
Mean mRSS reduced to 22.2 after 1 year and 18.5 at 4 years of follow-up from baseline 24.5. Mean FVC increased from 65% to 78.5% at 4 years follow-up, while DLCO increased from 55% to 77.73% No significant procedure-related side-effects

ASSIST: American scleroderma stem cell versus immune suppression trial, ASTIS: Autologous stem cell transplantation international scleroderma trial, CMV: Cytomegalovirus, DLCO: Diffusing capacity of the lungs for carbon monoxide, ESSG: European scleroderma study group, FEV1: Forced expiratory volume during first second, FVC: Forced vital capacity, G-CSF: Granulocyte-colony stimulating factor, HSCT: Hematopoietic stem cell transplantation, mRSS: modified Rodnan skin score, rATG: Rabbit antithymocyte globulin, PFT: Pulmonary function test, HRCT: High-resolution computed tomography, AEs: Adverse events

Patients with acute onset rapidly progressive disease refractory to conventional therapy and mild initial organ damage carry a better prognosis after hematopoietic stem cell therapy, while long standing disease, indolent course and irreversible organ damage are contraindications to this therapy.43 Hence, the challenge is to identify patients who are most likely to be benefitted, and the timing of hematopoietic stem cell therapy is to be tailored based on the phase of disease.

Risks of hematopoietic stem cell therapy include gonadal failure, secondary autoimmune diseases and malignancies. The European registries analyzed mortality after autologous hematopoietic stem cell therapy for severe autoimmune disease from 1996 to December 2007 and reported 5% mortality by day 100 following transplantation. The commonest cause of death included SSc recurrence, and transplant-related mortality, with others being cardiotoxicity, hemorrhage, secondary malignancies and infections.29 As systemic sclerosis has complex cardiac manifestations and cyclophosphamide is also associated with cardiotoxicity, a comprehensive pre-transplant cardiac assessment is recommended even in patients without cardiac symptoms.44 Hematopoietic stem cell therapy can induce gonadal failure in both sexes, therefore semen, oocyte or embryo cryopreservation/hormone replacement in case of gonadal failure should be considered as appropriate.45 The cumulative incidence of secondary autoimmune diseases was 9.8% after 5 years of treatment.46 In view of the high risk of treatment related side effects and early treatment-related mortality, the new EULAR treatment recommendations advise for careful selection of patients and highlight the experience of the medical team to be of utmost importance.47

Systemic Lupus Erythematosus

Hematopoietic stem cell therapy had been tried in patients with refractory systemic lupus erythematosus (SLE). The first case report of a successful autologous hematopoietic stem cell therapy in SLE was published in 1997, subsequent to which many observational studies and clinical trials have been conducted [Table 7].48-60 The current literature suggests that hematopoietic stem cell therapy is an option in refractory disease of short duration.61 European society for blood and marrow transplantation and British society of blood and marrow transplantation categorize autologous hematopoietic stem cell therapy in severe resistant disease as “clinical opinion,” that is, it can be undertaken after assessing risks and benefits.41 Guidelines from the American society for blood and marrow transplantation categorize the treatment as “developmental” in adults.42

Table 7: Studies on stem cell therapy in systemic lupus erythematosus
Author, year Indication and inclusion Methods Results Side effects
Marmont et al., 199748
Case report
A 48 year old woman, severe SLE since 20 years. Despite being on azathioprine, she required 40 mg of methylprednisolone to be in remission, recurrent chest pain (costochondritic) requiring intravenous corticosteroids Autologous BMT
Conditioning: 15mg/kg Thiotepa followed by 100 mg/kg of cyclophosphamide over 2 days
7 months after transplant, corticosteroid requirement reduced to 10 mg methylprednisolone daily, and ANA/anti-DNA antibodies not demonstrable. Chest pain did not recur Post-transplant course was uneventful
Burt et al., 199849
Case series,
2 patients
Patient 1: malar rash, arthralgias, hematuria, diffuse abdominal pain, pancytopenia ascites and pericardial effusion. Renal function was declining
Patient 2: active pneumonitis, pulmonary infiltrates, hypoxia, WHO class IV glomerulonephritis. Positive ANA
Source: bone marrow and peripheral blood stem cells
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide and antithymocyte globulin
Outcome: 50% improvement in serology, SLEDAI, serum creatinine, 24 h urine protein, creatinine clearance, left ventricular ejection fraction, chest radiograph and pulmonary function without any deterioration
Patient 1: At 12 months, malar rash, cytopenia, arthralgia, pericardial effusions resolved and renal function improved. Patient off all immunosuppressant medications
Patient 2: at 6 months follow-up, pulmonary infiltrates improved. PFTs and creatinine values static. ANA decreased. Steroids tapered from 80 mg/day to 25 mg/day
Cell lysis effect, acid base and electrolyte disorders, volume disturbances
Fouillard et al., 199950
Case report
A 35-year old woman with Raynaud’s phenomenon, arthritis, cutaneous vasculitis, proteinuria (WHO class III renal involvement) and ANA positivity (1:4096), anti-ds-DNA and anti-SSA antibodies, right homonymous hemianopia (left occipital ischemia) Autologous HSCT
Mobilization: cyclophosphamide and G-CSF
Conditioning regimen: BEAM
At 1-year follow-up, clinical remission – No Raynaud’s phenomenon, arthritis, cutaneous vasculitis. ANA and anti SSA negative at 1stand 6thmonth but become positive at 9 months. Renal - Reduced to class Ii Steroid requirement - 12.5 mg/day of prednisone Grade II mucositis
Brunner et al., 200251
Case report
Treatment resistant SLE with severe nephrotic syndrome and recurrent pneumonitis
Resistant to steroids, azathioprine, cyclophosphamide
Autologous HSCT
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: cyclophosphamide and ATG
At 21 months follow-up, disease under remission. Not on immunosuppressive drugs
CT chest - No SLE related pulmonary activity
Sepsis - Pseudomonas aeruginosa
Moderate intermittent fluid overload, mild mucositis
Lisukov et al., 200452
Case series,
6 patients
Recalcitrant SLE (cyclophosphamide pulse), with Class III or IV glomerulonephritis, CNS lupus, vasculitis involving the lung or heart, or life threatening cytopenias HSCT
Source: Bone marrow (4 patients) and peripheral (2 patients)
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: BEAM with ATG
Case 1: Complete remission. Steroid stopped at 10 months
Case 3: Improvement in CNS lupus. Low dose steroids (7.5–10 mg/day), azathioprine and cyclosporine
Case 6: Complete remission after 6 months
3 patients died. 1 - on day 11. Sepsis and multiple organ failure, hemorrhage
1 - on day 22. Sepsis followed by multiple organ failure, hemorrhage pneumonia
1 - on day 63. CMV infection (treated), pancytopenic. Sepsis and pneumonia followed by multiple organ failure
Burt et al, 200653
Case series,
49 patients
Patients with class III or IV glomerulonephritis or lung involvement (vasculitis, pneumonitis, alveolar hemorrhage), CNS involvement (cerebritis or transverse myelitis), vasculitis (biopsy proven or angiogram), biopsy proven myositis, transfusion-dependent autoimmune cytopenia, symptomatic pericardial or pleural effusions, ulcerative mucocutaneous disease, antiphospholipid syndrome refractory to treatment Autologous HSCT
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: Cyclophosphamide and equine antithymocyte globulin
At 5 years
Overall survival - 84%
Disease free survival - 50%
No remission - 4 patients
SLEDAI score improved and remained lower till 5 years
Creatinine clearance - Stable
Deaths: Disseminated mucormycosis before transplantation, but after stem cell mobilization - 1, non-treatment related - 6, SLE related - 4
Infections: Pneumocystis jiroveci - 1, esophageal candidiasis - 1, gram positive bacteremia - 4 during mobilization, 14 during transplantation; peritoneal fluid: Candida parapsilosis– 1; blood: Candida glabrata– 1; immune mediated hematological: acquired factor VIII deficiency – 2, ITP – 1
Gualandi et al., 200754
Case series,
8 patients (blood - 4, marrow - 1, mobilized stem cell - 3)
Severe SLE Autologous HSCT
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: Reduced intensity protocol thiotepa and cyclophosphamide
Complete remission following transplant
SLEDAI index improved from 90 to 9
Fever - All patients
Relapses - 2 patients
Loh et al., 200755
Case series,
13 patients
Mean SLEDAI - 20
Impaired left ventricular ejection fraction (6), pulmonary hypertension (5), mitral valve dysfunction (3), pericardial effusion (1)
Autologous HSCT
Mobilization: Cyclophosphamide and G-CSF
Conditioning regimen: Intravenous cyclophosphamide with equine ATG or alemtuzumab
Disease remission
Impaired LVEF - Stable or improved
Mitral valve disease improved
Elevated pulmonary
Pressures paralleled disease activity
5 patients - Fluid overload (mobilization or transplant course)
Deaths - 2 (post-transplant SLE progression). No cardiac events or transplant related deaths
Relapse - 2 patients
Vanikar et al., 200756
Retrospective cohort study,
27 patients
Biopsy proven lupus nephritis with high dsDNA antibodies, ANA and low serum C3 Allogenic HSCT
Mean follow-up 4.9 years
Average disease-free interval was 7.35 months (range, 2.1–12.7 months) No GVHD in any patient
Farge et al, 201029
Multi-centre observational study,
85 patients
All consecutive patients with autoimmune diseases who underwent stem cell transplantation Autologous HSCT
Mobilization: cyclophosphamide and G-CSF or G-CSF alone
Conditioning regimen: total body irradiation or single-agent chemotherapy or combinations based on cyclophosphamide, busulfan, and BEAM ± anti-thymocyte globulin
At 5 years, overall survival – 76%
Progression-free survival – 44%
At 100 days, transplant-related mortality – 11%
Alchi et al, 201257
Retrospective study,
28 patients
Patients reported to EBMT registry from 2001 to 2008 Autologous HSCT 5 years overall survival: 81±8%
Disease free survival: 29±9%
Nonrelapse mortality: 15±7%
Relapse incidence: 56±11%
Severe/life threatening AEs: 31 (15 - infections, 3 - severe immune events, 1 – post-transplant EBV associated lymphoproliferative disorder, secondary autoimmune disease - 2, cardiovascular events- 2)
Deaths: 5 (3 - infections, 1 - autoimmune hemolytic anemia, 1 - disease progression)
Leng et al., 201758
Prospective cohort study,
27 patients
Severe SLE (WHO class III or IV lupus nephritis progressive pulmonary dysfunction or pulmonary fibrosis, recurrent flares of lupus encephalopathy, transverse myelitis or catastrophic antiphospholipid syndrome) High-dose immunosuppressive therapy and autologous peripheral blood stem cell transplantation 21 (87.5%) cases achieved remission at 6 months 2 achieved partial remission and 1 patient died, 14 patients completed 10 years follow-up.
The 10-year overall survival rate and 10 years remission survival rate were both 86%, 16 (66.7%) patients remained in remission, four were lost to follow-up, two died, and one patient remained active
Infections (CMV, bacterial, both) - 12 (44.4%) patients, nausea, vomiting, alopecia, transaminitis
Burt et al., 201859
Prospective cohort study,
30 patients
Refractory SLE, steroid dependent, having organ damage, and at high risk of mortality Autologous HSCT using two different non-myeloablative conditioning regimens
four patients (Group 1): cyclophosphamide (200 mg/kg) and alemtuzumab (60 mg)
26 patients (Group 2): cyclophosphamide (200 mg/kg), rATG (Thymoglobulin) (5.5 mg/kg), and rituximab 1000 mg
Group 1: None entered remission
Group 2: Disease remission (defined as no immune suppressive drugs except hydroxychloroquine and/or ≤10 mg/day of prednisone) was 92% at 6 months, 92% at 1 year, 81% at 2 years, 71% at 3 years, and 62% at 4 and 5 years post-HSCT
Conclusion- Autologous HSCT outcome is dependent on conditioning regimen
No treatment related deaths
Infection - sinusitis (5), UTI (6), VZV (3), URTI (7), clostridium difficile (3), influenza (3), cellulitis (2)
Others, hip AVN, adrenal insufficiency, acute ITP
Cao et al., 201860
Prospective cohort study,
22 patients
SLE with failed previous therapy (prednisolone 0.5 mg/kg at least 2 months, or methylprednisolone pulse treatment for 6 months, or cyclophosphamide 500 mg/m2/month × 3 months) Autologous HSCT
Conditioning regimen of cyclophosphamide and ATG
3-year and 5 years
Progression-free survival was 77.27% and 67.9%, respectively
Overall survival rate was 95.2%. The titers of ANA, anti-dsDNA, anti-Sm antibody, and 24-h urinary protein significantly decreased, while complements 3 (C3) and C4 normalized at 100 days after transplantation (P<0.05)
Infections (virus, bacteria, PCP and TB)
The incidence of CMV reactivation was 59.09% post-transplantation in 3 years

ANA: Anti-nuclear antibody, AVN: Avascular necrosis, anti dsDNA: Anti-double stranded deoxyribonucleic acid, anti-Sm: Anti-smith antibody, anti-SSA: Anti–Sjögren’s-syndrome-related antigen A autoantibodies, BEAM: Carmustine, cytarabine, melphalan and etoposide, BMT: Bone marrow transplant, CMV: Cytomegalovirus, CNS: Central nervous system, EBMT: European group for blood and marrow transplantation, EBV: Epstein–Barr virus, GVHD: Graft versus host disease, CSF: Colony stimulating factor, G-CSF: Granulocyte-CSF, HSCT: Hematopoietic stem cell transplantation, ITP: Immune thrombocytopenic purpura, LVEF: Left ventricular ejection fraction, ATG: Antithymocyte antiglobulin, rATG: Rabbit ATG, PCP: Pneumocystis carinii pneumonia, SLE: Systemic lupus erythematosus, SLEDAI: SLE disease activity index, TB: Tuberculosis, URTI: Upper respiratory tract infection, UTI: Urinary tract infection, VZV: Varicella zoster virus, WHO: World Health Organization, PFTs: Pulmonary function tests, CT: Computed tomography, AEs: Adverse events

Therapeutic potential of mesenchymal stem cells has been explored in various autoimmune diseases including SLE.62 It has been found to be safe and effective in SLE, with decrease in disease activity, improvement in renal function, reduction in autoantibody production, peripheral Treg upregulation and re-establishment of balance between Th1- and Th2-related cytokines.63 Their immunomodulatory and regenerative characteristics make them a promising novel therapy for SLE.

Psoriasis

Although recent years have seen considerable progress in elucidating psoriasis pathogenesis, the exact mechanism is yet not fully known. At present, attention has been drawn to the possibility of dysfunction of certain types of stem cells to be the main cause of dysregulation of the inflammatory response in psoriasis.64 The idea originated while noticing psoriasis patients undergoing hematopoietic stem cell therapy and subsequently achieving long-term remission.65,66 In contrast, cases of acquired psoriasis after bone marrow transplantation from donors with psoriasis have also been reported.67-69 This suggests that hematopoietic stem cells have a significant role in disease pathogenesis. Mesenchymal stem cells have also been tried in few studies with success.70 Clinical benefits may be attributed to mesenchymal stem cell engraftment or to their paracrine or immunomodulatory effects. However, the availability of cost-effective, safe alternatives prevent the use of stem cell transplantation as a viable option in psoriasis.

Epidermolysis Bullosa

Although there is no specific treatment for this genetic condition till date, various therapeutic modalities are being studied that aim at correction of the underlying genetic defect and restore skin barrier. Stem cell therapy is one such cell based therapy. Either mesenchymal stem cells from the donor can be introduced intradermally or intravenously, bone marrow transplant can be done from allogeneic donor, or patient’s stem cells can be genetically modified and transplanted. While hematopoietic stem cell therapy has failed to live up to its initial promise, allogenic mesenchymal stem cells therapy may be useful in alleviating some symptoms.

In a study by Conget et al., two patients with severe generalized recessive dystrophic epidermolysis bullosa (EB) treated with intradermal administration of allogenic mesenchymal stem cells from bone marrow showed complete healing of ulcers around the treated site by 12 weeks.71 One week after intervention, type VII collagen was detected along the basement membrane zone and the dermal–epidermal junction was continuous in the treated site. However, the clinical effect lasted for only 4 months in both the patients.

A case of junctional EB treated with primary keratinocyte culture had normal morphology and absence of spontaneous and induced blisters or erosions at 21 months of follow-up.72

Studies by El-Darouti et al. and Wagner et al. using stem cells have also shown promising results.73,74 Petrof et al. studied ten recessive dystrophic EB children treated with intravenous allogeneic bone marrow-derived mesenchymal stem cells and found that the procedure was well tolerated and the adverse effects were minimal at nine months of follow-up.75 However, skin biopsy performed at day 60 showed no increase in type VII collagen and no new anchoring fibrils.

Although the initial clinical improvement was promising, it did not sustain with time due to lack of production of long lasting proteins (collagen and laminins). The current evidence of the use of stem cell therapy in EB is limited as the total number of patients treated with this modality is low, thus warranting further research to evaluate the efficacy and the potential risk to benefit correlation.76

Wound Healing

Epidermal stem cells have the potential to regenerate the epidermis and differentiate into various cell types and tissues, under appropriate stimuli.77 This property can be utilized to advantage in initiation and acceleration of healing of chronic non-healing wounds. Mesenchymal stem cells have been shown to promote wound healing by decreasing inflammation, promoting angiogenesis and decreasing scarring.78 Falanga et al. successfully applied human mesenchymal stem cells to non-healing and acute wounds, using a specialized fibrin spray system.79 Lu et al. demonstrated efficacy of stem cell therapy in diabetic foot ulcers.80

Vitiligo

Medical management is the first-line therapy for vitiligo and when it fails surgical therapies are considered.

Significant progress has been made in development of novel surgical techniques in vitiligo. The surgical modalities can be broadly divided into tissue and cellular grafts. Autologous non-cultured outer root sheath hair follicle cell suspension (NCORSHFS) is a recently described cellular graft technique. It is based on principle that hair follicle melanocytes have remarkable regenerative capacity which makes them a coveted source of melanocytes, instead of epidermis, for cell based therapies in vitiligo.81 Mohanty et al. in their pioneering study reported the use of NCORSHFS and reported a mean repigmentation of 65.7%.82 More than 75% repigmentation was achieved in 9/14 (64.2%) patients. The mean repigmentation rate was significantly less in patients with disease stability of less than 12 months duration. Vinay et al. studied the treatment variables determining therapeutic outcome in 30 patients with 60 target lesions undergoing NCORSHFS.83 Optimum repigmentation (> 75%) was seen in 21 of 60 (35%) lesions. The number of melanocytes (P = 0.04) and hair follicle stem cells (P = 0.01) transplanted was significantly higher among patients achieving optimum repigmentation. This, along with absence of dermal inflammation was significant predictors of achieving optimum repigmentation.

Moreover, multilineage-differentiating stress enduring (MUSE) cells are other type of stem cells that can have utility in treatment of vitiligo. Ex vivo studies in three-dimensional skin culture model have identified factors that induce MUSE cells to differentiate into melanocytes, which get integrated in the epidermis and lead to melanogenesis.84 The in vivo effect is yet to be determined.

Scleromyxedema

Scleromyxedema is a rare chronic fibro-mucinous disorder that may result in high mortality due to respiratory complications. Lacy et al. studied five patients who were given high-dose chemotherapy with stem cell rescue and found that it offers durable remission in most patients, although it is not curative.85 Illa et al. successfully treated scleromyxedema with chemotherapy and autologous stem cell transplantation.86 Full recovery was achieved at six months and the patient continued to be in remission three years after the transplantation.

Alopecia

Although application of stem cell therapy in hair restoration is relatively new and the stem cell isolation techniques are varied, the results to date are promising in both androgenetic alopecia (AGA) and alopecia areata (AA). Anderi et al. harvested autologous adipose-derived stromal vascular cells through lipoaspiration and injected into the scalp of 20 patients with AA.87 At three and six months of follow-up, they found statistically significant hair growth in all the patients. Adipose-derived stem cell conditioned medium (ADS-CM), known to be rich in growth factors such as vascular endothelial growth factor, hepatocyte growth factor, platelet-derived growth factor, and insulin-like growth factor ILGF, has also be utilized to treat hair loss.88 Gentile et al. isolated human adult stem cells by the centrifugation of human hair follicles obtained through punch biopsy and injected them into the scalps of 11 AGA patients resulting in an increase in hair density and count compared to baseline and placebo.89 Elmaadawi et al. who randomly assigned 40 patients (20 with AGA and 20 with AA) to receive either autologous bone marrow-derived mononuclear cells or autologous follicular stem cell injections into the scalp, found significant improvement in hair loss with no significant difference between the two preparations.90 Li et al. introduced a novel stem cell method, termed “stem cell educator therapy” in which patient’s mononuclear cells are separated from whole blood and allowed to interact with human cord blood–derived multipotent stem cells, and these “educated” cells are returned to patient’s circulation.91 Of nine patients with severe AA, all but one experienced improved hair regrowth of varying degrees. Two patients (one with alopecia totalis and one with patchy AA) experienced complete hair regrowth at 12 weeks without relapse after two years. A combination of platelet-rich plasma and stem cell technology has also shown promising results.92

Human Immunodeficiency Virus

Till date, there is no curative therapy for HIV. HIV binds to CD4 receptor, after which it needs CXCR4 or CCR5 as a co-receptor for gaining entry into the target cell. In 2009, Hütter et al. reported a case, known as the “Berlin patient”, of acute myeloid leukemia with HIV-1 infection who received allogenic HSCT twice from a donor with homozygous CCR5 delta32 allele. After transplantation, highly active anti-retroviral therapy (HAART) was stopped and the patient remained in remission at 20 months follow-up.93

Ten years later, another HIV patient of Hodgkin’s lymphoma underwent allogenic transplantation from donor with homozygous CCR5 delta32 allele following which antiretroviral therapy could be stopped after 16 months post-transplant and the patient was in remission after 18 months of stopping ART.94

Merkel Cell Carcinoma

It is a rare cutaneous tumor with no standard treatment protocol of metastatic disease. In recent years, there has been an increased understanding of Merkel cell carcinoma biology, especially with regard to the Merkel cell polyomavirus as the causative agent, suggesting that healthy human skin harbors resident or transient polyomavirus capable of neoplastic transformation. Significant differences between polyomavirus-positive and polyomavirus-negative Merkel cell carcinomas in terms of morphology, gene expression, signaling pathways, genomic and epigenetic alterations, microRNA profiles, dysregulated immune surveillance, aberrant protein expression and post-translational modifications have been reported, which influence the overall prognosis of Merkel cell carcinoma.95 Recent works suggest that polyomavirus-positive and negative Merkel cell carcinomas arise from two different cells of origin: the virus-negative carcinoma from epidermal stem cells and the virus-positive Merkel cell carcinoma from dermal stem cells.96 Further research on possible involvement of Merkel cell carcinoma stem cells could provide platform significant basis for preclinical development of targeted therapeutics. Anecdotal report of polychemotherapy with autologous peripheral blood stem cell transplantation resulted in remission, though it lasted only six months.97

Melanoma

Melanoma is an aggressive, relatively radio- and chemo-resistant tumor which is difficult to treat even with novel therapies including oncogene-directed therapy and immunotherapy. Recently, it has been suggested that cancer is a disease in which the persistence of the tumor relies on a small population of tumor-initiating cells, the tumor stem cells. This concept, though initially established for human myeloid leukemia, has been recently considered for melanoma (melanoma stem cells).98 These tumor stem cells are capable of self-renewal and thereby possess the ability for unlimited proliferation, are resistant to many therapeutic approaches and induce tumor relapse. Therefore, future therapies can be targeted at melanoma stem cell biomarkers, microenvironment and melanoma stem cells as a treatment option. Melanoma stem cells have been shown to express CD20 surface marker, ABCB5 (an ABC transporter protein), along with other markers including CD133, CD271 and aldehyde dehydrogenase.99 Since melanoma stem cells express CD20, rituximab, has been attempted in clinical trials to treat melanoma, producing regression in chemotherapy-refractory melanoma. Similarly, a monoclonal antibody against ABCB5+ melanoma stem cells in mouse models showed tumor inhibitory effects.100 Molecules involved in the mitogen-activated protein kinase (MAPK) signaling could represent interesting targets to overcome melanoma resistance as it has been shown that MAPK activation by mutant B-RAF drives melanoma tumor proliferation and that resistance to B-RAF inhibitors can be (or not) associated with reactivation of the MAPK pathway (“escape route”).101 The tumorigenic potential of tumor stem cells may be reduced by inhibition of essential stem cell factors or the therapeutic administration of differentiation factors.102

Aesthetic Medicine

Adipose-derived stem cells have been shown to activate fibroblasts and secrete various growth factors which lead to antioxidant, pigment lightening and wound-healing effects in the skin.103 They have been used effectively to treat wrinkles in animal models.104 Fibroblasts play a crucial role in wound healing and by their ability to restore lost dermal constituents; cultured autologous fibroblasts show promising future in aesthetic medicine.105

Current Status of Stem Cell Therapy in India

National Guidelines for Stem Cell Research, 2017, brought out by Indian Council of Medical Research (ICMR) and Department of Biotechnology (DBT), provides the basic guidelines needed for stem cell research in India.

These guidelines enumerate the various ethical issues, screening, categorization of research, levels of stem cell manipulation, stem cell research, manufacturing and release criteria for stem cells, procurement and banking, therapeutic uses, publicity and advertisements. The guidelines state that “The commercial use of stem cells as elements of therapy is prohibited. It must be emphasized that no stem cell administration to humans is permissible outside the purview of clinical trials”.106

At present, only hematopoietic stem cell therapy has been approved in India as per National Guidelines formulated by ICMR and DBT. According to the Ministry of Health and Family Welfare in 2017, there are 60 centers offering stem cell therapy in India. Western railway hospital, Vadodara, IKDRC, Ahmedabad and New Civil Hospital, Surat are the government institutions in the list of centers.107 The list of all the centers is included as an addendum.

According to the american society for blood and marrow transplantation, hematopoietic stem cell therapy is approved in about 31 diseases in adults and 43 diseases in children.42 The approved indications include hematological malignancies, solid tumors, non-malignant diseases (e.g., aplastic anemia, Fanconi’s anemia, dyskeratosis congenita and juvenile rheumatoid arthritis). Among dermatological diseases, the use of hematopoietic stem cell therapy had been approved in systemic sclerosis in children (<18 years of age) but not in adults by the Task force of ASBMT.

Difficulties in Stem Cell Therapy

  • Ethical considerations: These include issues of consent, confidentiality, screening for transmissible diseases and procedural risks.

  • Legal issues: There are many unauthorized centers in India that offer unproven stem cell therapies. The limitation is that, currently, the best available is only guidelines and not laws, due to which there is a gap between recommendation and implementation. A way to curb this practice is by making stringent rules and considering all stem cells and its products as drugs requiring drug trials before approval for a particular disease management.108

  • Possibility of graft rejection in allografting.

  • Tumorigenicity of pluripotent stem cells – Undesirable mutations may occur in stem cells when maintained for prolonged periods in culture and they may aberrantly differentiate to form tumors.

  • The consequence of preservation of stem cells or their products on their viability and potency must be assessed.

  • The quality control of cell processing and manufacturing must conform to the rules lain in Schedule M of Drugs and Cosmetics Act, 1940, which is sine qua non for all clinical trials.

  • Future research regarding ideal patient selection, timing of intervention, appropriate conditioning regimens, post-intervention care and cost effectiveness would help to optimize the results of stem cell therapy.

Declaration of patient consent

Patient’s consent not required as there are no patients in this study.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

  1. . Available from: http://nobelprize.org/nobel_prizes/medicine/laureates/2007/index.html [Last retrieved on 2007 Oct 08]
    [Google Scholar]
  2. . The Gazette of India: Extraordinary [Part ii-Sec. 3(i)] G.S.R. 227(E) In: New Drugs and Clinical Trials Rules. .
    [Google Scholar]
  3. , , . Plasticity of marrow-derived stem cells. Blood. 2003;102:3483-93.
    [CrossRef] [PubMed] [Google Scholar]
  4. . The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4:7-25.
    [Google Scholar]
  5. , , , , , . Stem cells: Sources and therapies. Biol Res. 2012;45:207-14.
    [CrossRef] [PubMed] [Google Scholar]
  6. , , , . Current and future perspectives of stem cell therapy in dermatology. Ann Dermatol. 2017;29:667-87.
    [CrossRef] [PubMed] [Google Scholar]
  7. , , . Culture systems for pluripotent stem cells. J Biosci Bioeng. 2005;100:12-27.
    [CrossRef] [PubMed] [Google Scholar]
  8. , , , . Niche interactions in epidermal stem cells. World J Stem Cells. 2015;7:495-501.
    [CrossRef] [PubMed] [Google Scholar]
  9. , , , , , . Bulge region as a putative hair follicle stem cells niche: A brief review. Iran J Public Health. 2017;46:1167-75.
    [Google Scholar]
  10. , , , , , , et al. Wnt/β-catenin and kit signaling sequentially regulate melanocyte stem cell differentiation in UVB-induced epidermal pigmentation. J Invest Dermatol. 2013;133:2753-62.
    [CrossRef] [PubMed] [Google Scholar]
  11. , , , , . Low-energy helium-neon laser induces locomotion of the immature melanoblasts and promotes melanogenesis of the more differentiated melanoblasts: Recapitulation of vitiligo repigmentation in vitro. J Invest Dermatol. 2006;126:2119-26.
    [CrossRef] [PubMed] [Google Scholar]
  12. . Dermabrasion may repigment vitiligo through stimulation of melanocyte precursors and elimination of hyperkeratosis. J Cosmet Dermatol. 2012;11:318-22.
    [CrossRef] [PubMed] [Google Scholar]
  13. , , , . FK506 (tacrolimus) and endothelin combined treatment induces mobility of melanoblasts: New insights into follicular vitiligo repigmentation induced by topical tacrolimus on sun-exposed skin. Br J Dermatol. 2011;164:490-6.
    [CrossRef] [PubMed] [Google Scholar]
  14. , . More than one way to skin. Genes Dev. 2008;22:976-85.
    [CrossRef] [PubMed] [Google Scholar]
  15. , , , , . Insights and hopes in umbilical cord blood stem cell transplantations. J Biomed Biotechnol. 2012;2012:572821.
    [CrossRef] [PubMed] [Google Scholar]
  16. , , . Corrigendum to “impact and challenges of mesenchymal stem cells in medicine: An overview of the current knowledge”. Stem Cells Int. 2019;2019:5493654.
    [CrossRef] [PubMed] [Google Scholar]
  17. , , , , , . Optimizing stem cells mobilization strategies to ameliorate patient outcomes: A review of guide-lines and recommendations. Int J Hematol Oncol Stem Cell Res. 2017;11:78-88.
    [Google Scholar]
  18. , . Conditioning regimens for hematopoietic cell transplantation: One size does not fit all. Blood. 2014;124:344-53.
    [CrossRef] [PubMed] [Google Scholar]
  19. , . Hematopoietic stem cell transplantation. Stem Cells Cloning. 2010;3:105-17.
    [CrossRef] [PubMed] [Google Scholar]
  20. , . Autologous stem cell mobilization and collection. Hematol Oncol Clin N Am. 2016;30:573-89.
    [CrossRef] [PubMed] [Google Scholar]
  21. , , , , , , et al. High-dose immune suppression and autologous hematopoietic stem cell transplantation in refractory pemphigus foliaceus. Bone Marrow Transplant. 2004;34:1097-8.
    [CrossRef] [PubMed] [Google Scholar]
  22. , , , , , . Pemphigus vulgaris treated with allogeneic hematopoietic stem cell transplantation following non-myeloablative conditioning. Eur Rev Med Pharmacol Sci. 2010;14:785-8.
    [Google Scholar]
  23. , , , , , . Allogenic hematopoietic stem cell transplantation in pemphigus vulgaris: A single-center experience. Indian J Dermatol. 2012;57:9-11.
    [CrossRef] [PubMed] [Google Scholar]
  24. , , , , , , et al. Application of autologous hematopoietic stem cell transplantation for pemphigus. Int J Dermatol. 2017;56:296-301.
    [CrossRef] [PubMed] [Google Scholar]
  25. , , , , , , et al. Haematopoietic stem cell transplantation for autoimmune diseases. Nat Rev Rheumatol. 2017;13:244-56.
    [CrossRef] [PubMed] [Google Scholar]
  26. , , , , , , et al. Autologous non-myeloablative hematopoietic stem cell transplantation in patients with systemic sclerosis. Bone Marrow Transplant. 2007;40:549-55.
    [CrossRef] [PubMed] [Google Scholar]
  27. , , , , , , et al. High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for severe systemic sclerosis: Long-term follow-up of the US multicenter pilot study. Blood. 2007;110:1388-96.
    [CrossRef] [PubMed] [Google Scholar]
  28. , , , , , , et al. Long-term follow-up results after autologous haematopoietic stem cell transplantation for severe systemic sclerosis. Ann Rheum Dis. 2008;67:98-104.
    [CrossRef] [PubMed] [Google Scholar]
  29. , , , , , , et al. Autologous hematopoietic stem cell transplantation for autoimmune diseases: An observational study on 12 years' experience from the European Group for Blood and Marrow Transplantation Working Party on Autoimmune Diseases. Haematologica. 2010;95:284-92.
    [CrossRef] [PubMed] [Google Scholar]
  30. , , , , , , et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): An open-label, randomised phase 2 trial. Lancet. 2011;378:498-506.
    [CrossRef] [Google Scholar]
  31. , , , , , , et al. Optimization of Autologous Stem Cell Transplantation for Systemic Sclerosis A single-center longterm experience in 26 patients with severe organ manifestations. J Rheumatol. 2012;39:269-75.
    [CrossRef] [PubMed] [Google Scholar]
  32. , , , , , , et al. Cardiac involvement and treatment-related mortality after nonmyeloablative haemopoietic stem-cell transplantation with unselected autologous peripheral blood for patients with systemic sclerosis: A retrospective analysis. Lancet. 2013;381:1116-24.
    [CrossRef] [Google Scholar]
  33. , , , , , , et al. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: A randomized clinical trial. JAMA. 2014;311:2490-8.
    [CrossRef] [PubMed] [Google Scholar]
  34. , , , , , , et al. Autologous stem cell transplantation with thiotepa-based conditioning in patients with systemic sclerosis and cardiac manifestations. Rheumatology (Oxford). 2014;53:919-22.
    [CrossRef] [PubMed] [Google Scholar]
  35. , , , , , , et al. Autologous hematopoietic stem cell transplantation has better outcomes than conventional therapies in patients with rapidly progressive systemic sclerosis. Bone Marrow Transplant. 2017;52:53-8.
    [CrossRef] [PubMed] [Google Scholar]
  36. , , , , , , et al. Myeloablative autologous stem-cell transplantation for severe scleroderma. N Engl J Med. 2018;378:35-47.
    [CrossRef] [PubMed] [Google Scholar]
  37. , , , , , , et al. Autologous haematopoietic stem cell transplantation for Japanese patients with systemic sclerosis: Long-term follow-up on a phase II trial and treatment-related fatal cardiomyopathy. Mod Rheumatol. 2018;28:879-84.
    [CrossRef] [PubMed] [Google Scholar]
  38. , , , , , . Stem cell transplant in systemic sclerosis: An Indian experience. Int J Rheum Dis. 2018;21:859-65.
    [CrossRef] [PubMed] [Google Scholar]
  39. , , . Haematopoietic stem cell transplantation in systemic sclerosis. RMD Open. 2018;4:e000533.
    [CrossRef] [PubMed] [Google Scholar]
  40. , , , , , , et al. Transplantation for autoimmune diseases in North and South America: A report of the Center for International Blood and Marrow Transplant Research. Biol Blood Marrow Transplant. 2012;18:1471-8.
    [CrossRef] [PubMed] [Google Scholar]
  41. Available from: http://bsbmt.org/indications-table/ [Last accessed on 2021 Feb 10]
    [Google Scholar]
  42. , , , , , , et al. Indications for autologous and allogeneic hematopoietic cell transplantation: Guidelines from the American society for blood and marrow transplantation. Biol Blood Marrow Transplant. 2015;21:1863-9.
    [CrossRef] [PubMed] [Google Scholar]
  43. , , , , . Autologous hematopoietic stem cell transplantation for treatment of systemic sclerosis. Front Immunol. 2018;9:2390.
    [CrossRef] [PubMed] [Google Scholar]
  44. , , , , , , et al. Cardiac involvement and treatment-related mortality after nonmyeloablative haemopoietic stem-cell transplantation with unselected autologous peripheral blood for patients with systemic sclerosis: A retrospective analysis. Lancet. 2013;381:1116-24.
    [CrossRef] [Google Scholar]
  45. , , , , , , et al. Haematopoietic SCT in severe autoimmune diseases: Updated guidelines of the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2012;47:770-90.
    [CrossRef] [PubMed] [Google Scholar]
  46. , , , , , , et al. Secondary autoimmune diseases occurring after HSCT for an autoimmune disease: A retrospective study of the EBMT autoimmune disease working party. Blood. 2011;118:1693-8.
    [CrossRef] [PubMed] [Google Scholar]
  47. , , , , , , et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76:1327-39.
    [CrossRef] [PubMed] [Google Scholar]
  48. , , , . Autologous marrow stem cell transplantation for severe systemic lupus erythematosus of long duration. Lupus. 1997;6:545-8.
    [CrossRef] [PubMed] [Google Scholar]
  49. , , , , , , et al. Treatment of autoimmune disease by intense immunosuppressive conditioning and autologous hematopoietic stem cell transplantation. Blood. 1998;92:3505-14.
    [CrossRef] [PubMed] [Google Scholar]
  50. , , , , , . Control of severe systemic lupus erythematosus after high-dose immunusuppressive therapy and transplantation of CD34+ purified autologous stem cells from peripheral blood. Lupus. 1999;8:320-3.
    [CrossRef] [PubMed] [Google Scholar]
  51. , , , , , , et al. Autologous blood stem cell transplantation in refractory systemic lupus erythematosus with severe pulmonary impairment: A case report: Autologous Stem Cell Transplantation in Refractory SLE. Arthritis Rheum. 2002;46:1580-4.
    [CrossRef] [PubMed] [Google Scholar]
  52. , , , , , , et al. High-dose immunosuppression with autologous stem cell transplantation in severe refractory systemic lupus erythematosus. Lupus. 2004;13:89-94.
    [CrossRef] [PubMed] [Google Scholar]
  53. , , , , , , et al. Nonmyeloablative hematopoietic stem cell transplantation for systemic lupus erythematosus. JAMA. 2006;295:527-35.
    [CrossRef] [PubMed] [Google Scholar]
  54. , , , , , , et al. Autologous stem cell transplantation for severe autoimmune diseases: A 10-year experience. Ann N Y Acad Sci. 2007;1110:455-64.
    [CrossRef] [PubMed] [Google Scholar]
  55. , , , , , , et al. Autologous hematopoietic stem cell transplantation in systemic lupus erythematosus patients with cardiac dysfunction: Feasibility and reversibility of ventricular and valvular dysfunction with transplant-induced remission. Bone Marrow Transplant. 2007;40:47-53.
    [CrossRef] [PubMed] [Google Scholar]
  56. , , , , , , et al. Hematopoietic stem cell transplantation in autoimmune diseases: The Ahmedabad experience. Transplant Proc. 2007;39:703-8.
    [CrossRef] [PubMed] [Google Scholar]
  57. , , , , , , et al. Autologous haematopoietic stem cell transplantation for systemic lupus erythematosus: Data from the European Group for Blood and Marrow Transplantation registry. Lupus. 2013;22:245-53.
    [CrossRef] [PubMed] [Google Scholar]
  58. , , , , , , et al. Good outcome of severe lupus patients with high-dose immunosuppressive therapy and autologous peripheral blood stem cell transplantation: A 10-year follow-up study. Clin Exp Rheumatol. 2017;35:494-9.
    [Google Scholar]
  59. , , , , , , et al. Five year follow-up after autologous peripheral blood hematopoietic stem cell transplantation for refractory, chronic, corticosteroid-dependent systemic lupus erythematosus: Effect of conditioning regimen on outcome. Bone Marrow Transplant. 2018;53:692-700.
    [CrossRef] [PubMed] [Google Scholar]
  60. , , , , , , et al. Erratum corrige: “Autologous peripheral blood haematopoietic stem cell transplantation for systemic lupus erythematosus:the observation of long-term outcomes in a Chinese centre” C Cao et al. Clin Exp Rheumatol. 2018;36:688.
    [Google Scholar]
  61. , . Is there a place for hematopoietic stem cell transplantation in rheumatology? Rheum Dis Clin North Am. 2019;45:399-416.
    [CrossRef] [PubMed] [Google Scholar]
  62. , . Mesenchymal stem cell therapy for autoimmune disease: Risks and rewards. Stem Cells Dev. 2015;24:2091-100.
    [CrossRef] [PubMed] [Google Scholar]
  63. . Therapeutic applications of mesenchymal stem cells for systemic lupus erythematosus. Adv Exp Med Biol. 2018;1089:73-85.
    [CrossRef] [PubMed] [Google Scholar]
  64. , , , , , . Stem cells as potential candidates for psoriasis cell-replacement therapy. Int J Mol Sci. 2017;18:2182.
    [CrossRef] [PubMed] [Google Scholar]
  65. , , , . Complete remission of both immunoglobulin light chain amyloidosis and psoriasis after autologous hematopoietic stem cell transplantation: A case report. Medicine (Baltimore). 2018;97:e13589.
    [CrossRef] [PubMed] [Google Scholar]
  66. , , , . Remission of psoriasis after allogeneic, but not autologous, hematopoietic stem-cell transplantation. J Am Acad Dermatol. 2013;68:489-92.
    [CrossRef] [PubMed] [Google Scholar]
  67. , , , , , . Psoriasis after allogeneic bone marrow transplantation. Arch Dermatol. 1990;126:1523.
    [CrossRef] [PubMed] [Google Scholar]
  68. , . Development of psoriasis after syngeneic bone marrow transplant from psoriatic donor: Further evidence for adoptive autoimmunity. Br J Dermatol. 1997;137:130-2.
    [CrossRef] [Google Scholar]
  69. , , , , , , et al. Transmission of psoriasis by allogeneic bone marrow transplantation and blood transfusion. Blood Cancer J. 2015;5:e288.
    [CrossRef] [PubMed] [Google Scholar]
  70. , , , , , , et al. Treatment of psoriasis with mesenchymal stem cells. Am J Med. 2016;129:e13-4.
    [CrossRef] [PubMed] [Google Scholar]
  71. , , , , , , et al. Replenishment of type VII collagen and re-epithelialization of chronically ulcerated skin after intradermal administration of allogeneic mesenchymal stromal cells in two patients with recessive dystrophic epidermolysis bullosa. Cytotherapy. 2010;12:429-31.
    [CrossRef] [PubMed] [Google Scholar]
  72. , , , , , , et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature. 2017;551:327-32.
    [CrossRef] [PubMed] [Google Scholar]
  73. , , , , , , et al. Treatment of dystrophic epidermolysis bullosa with bone marrow non-hematopoeitic stem cells: A randomized controlled trial. Dermatol Ther. 2016;29:96-100.
    [CrossRef] [PubMed] [Google Scholar]
  74. , , , , , , et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N Engl J Med. 2010;363:629-39.
    [CrossRef] [PubMed] [Google Scholar]
  75. , , , , , , et al. Potential of systemic allogeneic mesenchymal stromal cell therapy for children with recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 2015;135:2319-21.
    [CrossRef] [PubMed] [Google Scholar]
  76. , , , , , , et al. Progress towards treatment and cure of epidermolysis bullosa: Summary of the DEBRA International Research Symposium EB2015. J Invest Dermatol. 2016;136:352.e8.
    [CrossRef] [PubMed] [Google Scholar]
  77. , . Somatic epidermal stem cells can produce multiple cell lineages during development. Stem Cells. 2002;20:21-31.
    [CrossRef] [PubMed] [Google Scholar]
  78. , . Mesenchymal stem cells in chronic wounds: The spectrum from basic to advanced therapy. Adv Wound Care (New Rochelle). 2016;5:149-63.
    [CrossRef] [PubMed] [Google Scholar]
  79. , , , , , , et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng. 2007;13:1299-312.
    [CrossRef] [PubMed] [Google Scholar]
  80. , , , , , , et al. Comparison of bone marrow mesenchymal stem cells with bone marrow-derived mononuclear cells for treatment of diabetic critical limb ischemia and foot ulcer: A double-blind, randomized, controlled trial. Diabetes Res Clin Pract. 2011;92:26-36.
    [CrossRef] [PubMed] [Google Scholar]
  81. , , . Hair follicle renewal: Authentic morphogenesis that depends on a complex progression of stem cell lineages. Development. 2010;137:569-77.
    [CrossRef] [PubMed] [Google Scholar]
  82. , , , , . Noncultured extracted hair follicle outer root sheath cell suspension for transplantation in vitiligo. Br J Dermatol. 2011;164:1241-6.
    [CrossRef] [PubMed] [Google Scholar]
  83. , , , , , , et al. Clinical and treatment characteristics determining therapeutic outcome in patients undergoing autologous non-cultured outer root sheath hair follicle cell suspension for treatment of stable vitiligo. J Eur Acad Dermatol Venereol. 2015;29:31-7.
    [CrossRef] [PubMed] [Google Scholar]
  84. , , , , , , et al. Pluripotent nontumorigenic multilineage differentiating stress enduring cells (Muse cells): A seven-year retrospective. Stem Cell Res Ther. 2017;8:227.
    [CrossRef] [PubMed] [Google Scholar]
  85. , , , , , , et al. Successful treatment of scleromyxedema with autologous peripheral blood stem cell transplantation. Arch Dermatol. 2005;141:1277-82.
    [CrossRef] [PubMed] [Google Scholar]
  86. , , , , , , et al. Steady remission of scleromyxedema 3 years after autologous stem cell transplantation: An in vivo and in vitro study. Blood. 2006;108:773-4.
    [CrossRef] [PubMed] [Google Scholar]
  87. , , , , . Cellular therapy with human autologous adipose-derived adult cells of stromal vascular fraction for alopecia areata. Stem Cell Res Ther. 2018;9:141.
    [CrossRef] [PubMed] [Google Scholar]
  88. , , , , . Clinical use of conditioned media of adipose tissue-derived stem cells in female pattern hair loss: A retrospective case series study. Int J Dermatol. 2015;54:730-5.
    [CrossRef] [PubMed] [Google Scholar]
  89. , , , , . Stem cells from human hair follicles: First mechanical isolation for immediate autologous clinical use in androgenetic alopecia and hair loss. Stem Cell Investig. 2017;4:58.
    [CrossRef] [PubMed] [Google Scholar]
  90. , , , , , , et al. Stem cell therapy as a novel therapeutic intervention for resistant cases of alopecia areata and androgenetic alopecia. J Dermatolog Treat. 2018;29:431-40.
    [CrossRef] [PubMed] [Google Scholar]
  91. , , , , , , et al. Hair regrowth in alopecia areata patients following Stem Cell Educator therapy. BMC Med. 2015;13:87.
    [CrossRef] [PubMed] [Google Scholar]
  92. , , . Introducing Platelet-Rich Stroma: Platelet-Rich Plasma (PRP) and Stromal Vascular Fraction (SVF) Combined for the Treatment of Androgenetic Alopecia. Aesthet Surg J. 2018;38:811-22.
    [CrossRef] [PubMed] [Google Scholar]
  93. , , , , , , et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med. 2009;360:692-8.
    [CrossRef] [PubMed] [Google Scholar]
  94. , , , , , , et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature. 2019;568:244-8.
    [CrossRef] [PubMed] [Google Scholar]
  95. . Viral etiology of merkel cell carcinoma: Implications in diagnosis, prognosis, therapy and prevention. Int J Clin Dermatol Res. 2015;3:1-2.
    [CrossRef] [Google Scholar]
  96. , , , . Are there multiple cells of origin of Merkel cell carcinoma? Oncogene. 2018;37:1409-16.
    [CrossRef] [PubMed] [Google Scholar]
  97. , , , , , , et al. Transient complete remission of metastasized Merkel cell carcinoma by high-dose polychemotherapy and autologous peripheral blood stem cell transplantation. Br J Dermatol. 2000;143:837-9.
    [CrossRef] [PubMed] [Google Scholar]
  98. , , , , , . Stem cells, melanoma and cancer stem cells: The good, the bad and the evil? G Ital Dermatol Venereol. 2009;144:287-96.
    [Google Scholar]
  99. , , . Melanoma stem cells: The past, present and future. J Stem Cell Res Ther. 2018;4:89-90.
    [CrossRef] [Google Scholar]
  100. , , , , , , et al. Identification of cells initiating human melanomas. Nature. 2008;451:345-9.
    [CrossRef] [PubMed] [Google Scholar]
  101. . Latest approved therapies for metastatic melanoma: What comes next? J Skin Cancer. 2013;2013:735282.
    [CrossRef] [PubMed] [Google Scholar]
  102. , , , , , , et al. Melanoma stem cells: Targets for successful therapy? J Dtsch Dermatol Ges. 2008;6:541-6.
    [CrossRef] [PubMed] [Google Scholar]
  103. , , , , . Adipose-derived stem cells as a new therapeutic modality for ageing skin. Exp Dermatol. 2011;20:383-7.
    [CrossRef] [PubMed] [Google Scholar]
  104. , , , , . Antiwrinkle effect of adipose-derived stem cell: Activation of dermal fibroblast by secretory factors. J Dermatol Sci. 2009;53:96-102.
    [CrossRef] [PubMed] [Google Scholar]
  105. , . Role of cultured skin fibroblasts in aesthetic and plastic surgery. World J Plast Surg. 2013;2:2-5.
    [Google Scholar]
  106. . . Available from: http://www.icmr.nic.in/guidelines/Guidelines_for_stem_cell_research_2017.pdf [Last accessed on 2019 Dec 26]
    [Google Scholar]
  107. . Government of India Ministry of Health and Family Welfare, Lok Sabha, Unstarred Question No. 3448. Published Online 2017
    [Google Scholar]
  108. , . Unproven stem cell therapies in India: Regulatory challenges and proposed paths forward. Cell Stem Cell. 2018;23:649-52.
    [CrossRef] [PubMed] [Google Scholar]
Show Sections