• Low-dose IL-2 is safe and well tolerated at variable dosing regimens in pediatric patients with steroid-refractory chronic GVHD.

  • Low-dose IL-2 is effective in combination with other immunosuppressive medications.

The majority of patients with chronic graft-versus-host disease (cGVHD) are steroid refractory (SR), creating a need for safe and effective therapies. Subcutaneous low-dose interleukin-2 (LD IL-2), which preferentially expands CD4+ regulatory T cells (Tregs), has been evaluated in 5 clinical trials at our center with partial responses (PR) in ∼50% of adults and 82% of children by week 8. We now report additional real-world experience with LD IL-2 in 15 children and young adults. We conducted a retrospective chart review of patients with SR-cGVHD at our center who received LD IL-2 from August 2016 to July 2022 not on a research trial. The median age at start of LD IL-2 was 10.4 years (range, 1.2-23.2 years) at a median of 234 days from cGVHD diagnosis (range, 11-542 days). Patients had a median of 2.5 (range, 1-3) active organs at LD IL-2 start and received a median of 3 (range, 1-5) prior therapies. The median duration of LD IL-2 therapy was 462 days (range, 8-1489 days). Most patients received 1 × 106 IU/m2 per day. There were no serious adverse effects. The overall response rate in 13 patients who received >4 weeks of therapy was 85% (complete response, n = 5; PR, n = 6) with responses in diverse organs. Most patients significantly weaned corticosteroids. Tregs preferentially expanded with a median peak fold increase of 2.8 in the ratio of Tregs to CD4+ conventional T cells (range, 2.0-19.8) by 8 weeks on therapy. LD IL-2 is a well-tolerated, steroid-sparing agent with a high response rate in children and young adults with SR-cGVHD.

Chronic graft-versus-host disease (cGVHD) is a multisystem disorder that complicates allogeneic hematopoietic cell transplantation (HCT) in between 20% and 50% of children.1 It arises from a failure of donor-derived B and T cells to develop tolerance to allo- and autoantigens during immune reconstitution after transplantation.1,2 The immune-mediated damage in cGVHD eventually results in fibrotic changes that can affect any organ system and can lead to debilitating complications such as sclerotic skin, joint contractures, and bronchiolitis obliterans syndrome. Given these serious multiorgan effects, cGVHD is the leading cause of nonrelapse morbidity and mortality after allogeneic HCT.1,2 

The development of cGVHD is driven by tissue damage that primes dendritic cells to display costimulatory molecules to T cells.3 Subsequently, allo- and autoreactive T cells can activate pathogenic B cells, which release damaging immunoglobulins. Lymphocyte activation is accompanied by macrophage activation and release of profibrotic growth factors.3 Because of thymic injury from transplant conditioning and T-cell alloreactivity, central tolerance is impaired. Poor reconstitution of the CD4+ regulatory T-cell (Treg) compartment after transplantation also leads to dysfunctional peripheral tolerance.3-7 The failures in central and peripheral tolerance enable unchecked inflammation to drive fibrosis.

With broad immunosuppressive effects, corticosteroids remain the first-line therapy for cGVHD8; however, prolonged steroid exposure is associated with significant morbidity, and between 40% and 50% of patients are steroid refractory (SR).9 There are now 3 US Food and Drug Administration (FDA)-approved second-line agents for treatment of refractory cGVHD: ibrutinib, ruxolitinib, and belumosudil, with overall response rates (ORR) of 50% to 77% in phase 2 and 3 adult studies.10,11 Ibrutinib inhibits Bruton tyrosine kinase and IL-2 inducible T-cell kinase, targeting pathogenic B- and T-cell activation, respectively. Ruxolitinib is a Janus kinase (JAK) inhibitor that blocks signaling through multiple cytokine receptors on lymphocytes. Finally, belumosudil inhibits Rho-associated coiled coil–containing protein kinase-2, which prevents STAT3 phosphorylation and development of Th17 and T-follicular helper cell populations.3 

An alternative approach to refractory cGVHD developed at our center over the last 15 years is to augment the Treg compartment using low-dose interleukin-2 (LD IL-2). IL-2 is an essential cytokine for the proliferation and function of both regulatory and effector T cells. However, only Tregs constitutively express the higher affinity trimeric αβγ IL-2 receptor, enabling the preferential expansion of Tregs with low doses of IL-2.12 There have been 5 clinical trials investigating daily subcutaneous injections of LD IL-2 for SR-cGVHD at the Dana-Farber Cancer Institute (NCT00529035, NCT01366092, NCT01937468, NCT02318082, and NCT02340676), one of which included pediatric patients.13-18 In this phase 1 intrapatient dose-escalation study (NCT02318082), pediatric patients tolerated the previously defined maximum tolerated dose in adults of 1 × 106 IU/m2 per day. By 8 weeks, 82% (9 of 11) of children attained a partial response (PR) and 2 patients attained a complete response (CR) on extended-duration therapy. This ORR was higher than the 50% ORR typically observed among adult participants in our LD IL-2 trials. As a result, LD IL-2 is now offered as an off-trial second-line option for pediatric patients with SR-cGVHD at our center.

Data from clinical trials are often complemented by observations from real-world clinical use, which is less restrictive and more generalizable to diverse settings. To date, to the best of our knowledge, there are no reports of such data for LD IL-2 in pediatric patients for any disease indication. Here, we present our experience using LD IL-2 off study in pediatric and young adult patients with SR-cGVHD.

Patient selection

We conducted a retrospective chart review of all patients who received LD IL-2 for SR-cGVHD at the Dana-Farber/Boston Children’s Cancer and Blood Disorders Center while not on a clinical trial between August 2016 and July 2022. We collected patient demographic and transplant-related data via a data pull from Ottr (CareDx), an institutional data repository for patients who have received HCT. Race and ethnicity data recorded in this database are per patient self-report. We collected data for cGVHD involvement and severity, medication exposures, infectious or other complications, and disease response by manual chart review. Patients who received <4 weeks of therapy were considered inevaluable for clinical response based on experience from our prior studies in which clinical benefit required at least 8 to 12 weeks of therapy.13-18 This study was approved by the Dana-Farber/Harvard Cancer Center institutional review board (DFCI 21-417) and conducted in accordance with the Declaration of Helsinki.

LD IL-2 preparation

Recombinant IL-2 (aldesleukin; Clinigen) was provided as a lyophilized powder in vials of 22 million IU. Vials were aseptically diluted at the Dana-Farber Cancer Institute pharmacy by qualified personnel in a laminar flow hood with 1.2 mL sterile water and 4.8 mL dextrose 5% in water, resulting in a final IL-2 concentration of 3.67 million IU/mL. After reconstitution, a 14-day supply was provided to families in single-use syringes for home refrigerator storage at 2 to 8°C.

Clinical assessments

We determined clinical response and potential adverse events related to LD IL-2 by reviewing clinic visit and stem cell transplant inpatient notes in the electronic medical record, starting the month before LD IL-2 initiation through the end date of LD IL-2 therapy. Two to 3 physicians (H.W., J.S.W., and M.K.) adjudicated the response for each patient, and responses were based on 2014 National Institutes of Health (NIH) consensus criteria.19,20 

Immune cell analysis

Cell counts and lymphocyte populations data were collected as available in the medical record. Flow cytometry subsets performed through the Boston Children’s Hospital clinical laboratory included CD19+ cells (B cells), CD3+ cells (T cells) with CD4+ and CD8+ subsets, as well as CD4+ CD25+ CD127low cells (CD4+ Tregs). Natural killer (NK) cells were identified as CD3 and either CD16+ or CD56+.

Statistical analysis

Data analysis was primarily descriptive. The Wilcoxon signed-rank test was used for paired comparison, and multiple comparisons were not considered. Statistical analyses were conducted using GraphPad Prism version 9 (San Diego) and R version 4.2.2 (the CRAN project).

Patient characteristics

Fifteen patients received LD IL-2 for SR-cGVHD off study at the Dana-Farber/Boston Children’s Cancer and Blood Disorders Center between August 2016 and July 2022. This cohort included 2 patients (patient [P]7 and P12) who restarted, or continued, LD IL-2 therapy off study after initially being treated on the phase 1 trial (NCT02318082). Baseline patient characteristics are shown in Table 1. Seventy-three percent (11 of 15) of patients were male; 53% (8 of 15) of patients self-identified as White. The median age at stem cell transplantation was 9.1 years (range, 0.3-21.4 years). Fifty-three percent (8 of 15) of patients underwent transplantation for nonmalignant indications, 67% (10 of 15) received myeloablative conditioning, 40% (6 of 15) had mismatched unrelated donors, and 87% (13 of 15) received bone marrow as the stem cell source. Baseline transplant details were not available for 1 patient (P15) who received her stem cell transplant in another country.

Table 1.

Patient baseline characteristics

Number (N = 15)Percentage (%)
Age at transplant (y), median (range) 9.1 (0.3-21.4) 
Sex 
Male 11 73% 
Female 27% 
Race 
White 53% 
Black 13% 
Asian/Pacific Islander 7% 
Other 20% 
Not disclosed 7% 
Ethnicity 
Non-Hispanic/Latino 53% 
Hispanic/Latino 27% 
Not disclosed 20% 
Diagnosis   
Nonmalignant 53% 
Malignant 47% 
Conditioning 
MAC 10 67% 
RIC 20% 
None 7% 
Unknown  7% 
HLA type (A, B, C, DRB1) 
Matched unrelated 33% 
Matched related 20% 
Mismatched unrelated 40% 
Haploidentical 7% 
Cell source 
Bone marrow 13 87% 
Peripheral blood 7% 
Unknown  7% 
PrioraGVHD 
Yes 60% 
No 33% 
Unknown  7% 
Time to cGVHD from HCT (d), median (range) 168 (100-420) 
Time to LD IL-2 from cGVHD diagnosis (d), median (range) 234 (11-542) 
Age at LD IL-2 start (y), median (range) 10.4 (1.2-23.2) 
Time on LD IL-2 (d), median (range) 462 (8-1489) 
Number (N = 15)Percentage (%)
Age at transplant (y), median (range) 9.1 (0.3-21.4) 
Sex 
Male 11 73% 
Female 27% 
Race 
White 53% 
Black 13% 
Asian/Pacific Islander 7% 
Other 20% 
Not disclosed 7% 
Ethnicity 
Non-Hispanic/Latino 53% 
Hispanic/Latino 27% 
Not disclosed 20% 
Diagnosis   
Nonmalignant 53% 
Malignant 47% 
Conditioning 
MAC 10 67% 
RIC 20% 
None 7% 
Unknown  7% 
HLA type (A, B, C, DRB1) 
Matched unrelated 33% 
Matched related 20% 
Mismatched unrelated 40% 
Haploidentical 7% 
Cell source 
Bone marrow 13 87% 
Peripheral blood 7% 
Unknown  7% 
PrioraGVHD 
Yes 60% 
No 33% 
Unknown  7% 
Time to cGVHD from HCT (d), median (range) 168 (100-420) 
Time to LD IL-2 from cGVHD diagnosis (d), median (range) 234 (11-542) 
Age at LD IL-2 start (y), median (range) 10.4 (1.2-23.2) 
Time on LD IL-2 (d), median (range) 462 (8-1489) 

aGVHD, acute GVHD; MAC, myeloablative conditioning; RIC, reduced intensity conditioning.

One patient had received transplantation elsewhere with few records before her presentation for evaluation of her cGVHD.

cGVHD characteristics

The median time from HCT to cGVHD diagnosis was 168 days (range, 100-420 days), and the median time from cGVHD to LD IL-2 initiation was 234 days (range, 11-542 days). The median age at the start of LD IL-2 treatment was 10.4 years (range, 1.2-23.2 years). Patients tried a median of 3 (range, 1-5) therapies (including steroids) before IL-2 and had a median of 2.5 (range, 1-3) actively affected organs at the time of IL-2 treatment initiation. All patients had moderate-to-severe cGVHD per 2014 NIH consensus criteria.19 Individual patient characteristics including transplant indication, comorbidities, cGVHD features, and response to LD IL-2 are provided in Table 2.

Table 2.

Details on individual patients and IL-2 course

Transplant indicationcGVHD featuresPrevious GVHD therapies Other previous comorbiditiesAge at LD IL-2 start (y)Clinical response (excluding oral/ocular)Adverse events during therapy (not thought to be from LD IL-2)Reason for IL-2 dose change or discontinuation
P1 Fanconi anemia Skin
Ocular 
Steroid
Sirolimus 
  • Acute GVHD: gut

 
10.6 Overall CR
Skin CR 
 cGVHD stable/improved, thus weaned off therapy 
P2 AML Lung
JMF 
Steroid
Imatinib
Ruxolitinib 
 18.1 Overall PR
Lung SD
JMF PR 
Pulmonary aspergillosis Reduced to 0.33 dosing because of pain related to injections
Stopped because of pulmonary aspergillosis 
P3 Mixed leukemia Liver
Ocular
Lung  
Steroid
Ruxolitinib
Vedolizumab 
  • Acute GVHD: skin, oral, gut, liver

  • Stem cell boost for low CD34 counts

  • Eosinophilic gastroenteritis

 
18.4 Overall PR
Liver CR
Lung SD 
Pulmonary aspergillosis with development of pulmonary GVHD 6.5 mos break after aspergillosis; then restarted LD IL-2; cGVHD stable/improved, thus weaned off therapy 
P4 Immune deficiency (WAS) Lung Steroid 
  • Initial graft failure followed by second transplant

  • Chronic relapsing encephalomyelitis

 
4.3 Overall PR
Lung PR 
 cGVHD stable/improved, thus weaned off therapy 
P5 fHLH (STXBP2) Skin
Gut
Ocular 
Steroid
Tacrolimus
Vedolizumab,
Ruxolitinib  
  • Acute GVHD: skin, gut

  • Recurrent CVL infection

  • Waning chimerism → stem cell boost

 
4.6 Overall CR
Skin CR
Gut (not active at time of start) 
 cGVHD stable/improved, thus weaned off therapy 
P6 Omenn syndrome (IL-7R deficiency) Skin
Gut 
Steroid 
  • Acute GVHD: gut

  • Feeding intolerance; on parenteral nutrition

  • Central sleep apnea

  • Diabetes

  • CMV and EBV viremia

  • Tracheostomy with recurrent tracheitis and bacteremia

 
1.2 Overall PR
Skin CR
Gut PR 
Idiopathic giant cell myocarditis Held during infections & cardiac workup (2 mo)
IL-2 eventually paused for trial of mepolizumab (unclear if there would be an interaction) 
P7 Immune deficiency (unknown) Lung Steroid
Cyclosporine
MMF 
  • Central nervous system EBV+ leiomyosarcoma (resected ×3)

  • Chronic CMV viremia

  • Toxic epidermal necrolysis

 
8.4 Overall SD
Lung SD 
Influenza with multifocal bacterial pneumonia Was initially enrolled on pediatric cohort of LD IL-2 trial and had PR in lungs at week 8. Came off trial at week 97 because of progression in setting of respiratory infection
Restarted IL-2 off study 2 mos later. cGVHD stable/improved, thus weaned off therapy 
P8 AML Liver
Ocular
Oral 
Steroid
Ibrutinib,
Infliximab  
  • Second transplant after initial relapse of AML

  • Severe malnutrition

  • Organizing pneumonia

 
18.4 Inevaluable  50% dosing for prerenal AKI
Severe liver failure at time of IL-2 start; goals of care soon redirected to comfort measures 
P9 ALD Liver
Ocular
JMF 
Steroid
Cyclosporine
Rituximab,
Ibrutinib,
Ruxolitinib  
 10.2 Overall CR
Liver CR
JMF CR 
 Fatigue & flushing; switched to every-other-day dosing; cGVHD stable/improved, thus weaned off therapy 
P10 AML Lung
Skin  
Steroid
Cyclosporine
Ruxolitinib 
  • Acute GVHD: skin

  • Pulmonary superinfection & scarring

  • HHV-6 & BK Virus

 
12.7 Overall SD
Lung SD 
Pseudomonas pneumonia Insurance stopped covering 
P11 SAA Skin
Liver
Lung 
Steroid
Cyclosporine
Ruxolitinib 
  • Acute GVHD: liver, gut

  • Thrombocytopenia

 
10.2 Inevaluable  Stopped IL-2 & ruxolitinib because of thrombocytopenia; later the cause was determined to be immune thrombocytopenia 
P12 Gamma-delta
T-cell lymphoma 
Skin
Liver
Lung 
Steroid 
  • Acute GVHD: liver, gut, skin

  • Polymicrobial infected hemorrhagic pancreatic pseudocyst → distal pancreatectomy, splenectomy, & near-total colectomy

  • Steroid-induced chronic kidney disease

 
23.2 Overall CR
Skin CR
Liver CR
Lung CR 
Pancreatitis On study for 122 weeks and then continued off-study for 90 weeks cGVHD stable/improved, thus weaned off therapy 
P13 Immune deficiency (ITK deficiency) Skin
Gut 
Steroid
Tacrolimus
Vedolizumab 
  • Acute GVHD: skin

  • Liver transplant before HCT for genetic liver failure; complicated by wound dehiscence and intra-abdominal infection requiring abdominal wash out and biliary duct dilation requiring ERCP and stent placement in the common bile duct.

  • Poor graft function despite stem cell boost

  • Persistent HHV-6 viremia

  • Chronic pericardial effusion

 
4.1 Overall CR
Skin CR
Gut CR 
Bacteremia Given persistent graft failure, infections, and multiorgan dysfunction, switched to comfort care 
P14 B-ALL Skin
Liver
Oral 
Steroid
MMF 
  • Acute GVHD: skin

 
10.4 Overall PR
Skin SD
Liver PR 
 Mild improvement in skin but early plateau; added ruxolitinib & stopped IL-2 
P15 Myeloproliferative disorder (JAK2 mutation) Skin
Liver
Ocular 
Steroid
Ruxolitinib 
  • Acute GVHD status unknown

  • Bilateral breast tissue ulceration/sclerodermatous change with MDR PsA that preceded starting IL-2

 
15.9 Overall PR
Skin PR
Liver CR 
Recurrence of breast tissue infection Malaise at daily dosing, switched to every other day
Discontinued, because unavailable in home country 
Transplant indicationcGVHD featuresPrevious GVHD therapies Other previous comorbiditiesAge at LD IL-2 start (y)Clinical response (excluding oral/ocular)Adverse events during therapy (not thought to be from LD IL-2)Reason for IL-2 dose change or discontinuation
P1 Fanconi anemia Skin
Ocular 
Steroid
Sirolimus 
  • Acute GVHD: gut

 
10.6 Overall CR
Skin CR 
 cGVHD stable/improved, thus weaned off therapy 
P2 AML Lung
JMF 
Steroid
Imatinib
Ruxolitinib 
 18.1 Overall PR
Lung SD
JMF PR 
Pulmonary aspergillosis Reduced to 0.33 dosing because of pain related to injections
Stopped because of pulmonary aspergillosis 
P3 Mixed leukemia Liver
Ocular
Lung  
Steroid
Ruxolitinib
Vedolizumab 
  • Acute GVHD: skin, oral, gut, liver

  • Stem cell boost for low CD34 counts

  • Eosinophilic gastroenteritis

 
18.4 Overall PR
Liver CR
Lung SD 
Pulmonary aspergillosis with development of pulmonary GVHD 6.5 mos break after aspergillosis; then restarted LD IL-2; cGVHD stable/improved, thus weaned off therapy 
P4 Immune deficiency (WAS) Lung Steroid 
  • Initial graft failure followed by second transplant

  • Chronic relapsing encephalomyelitis

 
4.3 Overall PR
Lung PR 
 cGVHD stable/improved, thus weaned off therapy 
P5 fHLH (STXBP2) Skin
Gut
Ocular 
Steroid
Tacrolimus
Vedolizumab,
Ruxolitinib  
  • Acute GVHD: skin, gut

  • Recurrent CVL infection

  • Waning chimerism → stem cell boost

 
4.6 Overall CR
Skin CR
Gut (not active at time of start) 
 cGVHD stable/improved, thus weaned off therapy 
P6 Omenn syndrome (IL-7R deficiency) Skin
Gut 
Steroid 
  • Acute GVHD: gut

  • Feeding intolerance; on parenteral nutrition

  • Central sleep apnea

  • Diabetes

  • CMV and EBV viremia

  • Tracheostomy with recurrent tracheitis and bacteremia

 
1.2 Overall PR
Skin CR
Gut PR 
Idiopathic giant cell myocarditis Held during infections & cardiac workup (2 mo)
IL-2 eventually paused for trial of mepolizumab (unclear if there would be an interaction) 
P7 Immune deficiency (unknown) Lung Steroid
Cyclosporine
MMF 
  • Central nervous system EBV+ leiomyosarcoma (resected ×3)

  • Chronic CMV viremia

  • Toxic epidermal necrolysis

 
8.4 Overall SD
Lung SD 
Influenza with multifocal bacterial pneumonia Was initially enrolled on pediatric cohort of LD IL-2 trial and had PR in lungs at week 8. Came off trial at week 97 because of progression in setting of respiratory infection
Restarted IL-2 off study 2 mos later. cGVHD stable/improved, thus weaned off therapy 
P8 AML Liver
Ocular
Oral 
Steroid
Ibrutinib,
Infliximab  
  • Second transplant after initial relapse of AML

  • Severe malnutrition

  • Organizing pneumonia

 
18.4 Inevaluable  50% dosing for prerenal AKI
Severe liver failure at time of IL-2 start; goals of care soon redirected to comfort measures 
P9 ALD Liver
Ocular
JMF 
Steroid
Cyclosporine
Rituximab,
Ibrutinib,
Ruxolitinib  
 10.2 Overall CR
Liver CR
JMF CR 
 Fatigue & flushing; switched to every-other-day dosing; cGVHD stable/improved, thus weaned off therapy 
P10 AML Lung
Skin  
Steroid
Cyclosporine
Ruxolitinib 
  • Acute GVHD: skin

  • Pulmonary superinfection & scarring

  • HHV-6 & BK Virus

 
12.7 Overall SD
Lung SD 
Pseudomonas pneumonia Insurance stopped covering 
P11 SAA Skin
Liver
Lung 
Steroid
Cyclosporine
Ruxolitinib 
  • Acute GVHD: liver, gut

  • Thrombocytopenia

 
10.2 Inevaluable  Stopped IL-2 & ruxolitinib because of thrombocytopenia; later the cause was determined to be immune thrombocytopenia 
P12 Gamma-delta
T-cell lymphoma 
Skin
Liver
Lung 
Steroid 
  • Acute GVHD: liver, gut, skin

  • Polymicrobial infected hemorrhagic pancreatic pseudocyst → distal pancreatectomy, splenectomy, & near-total colectomy

  • Steroid-induced chronic kidney disease

 
23.2 Overall CR
Skin CR
Liver CR
Lung CR 
Pancreatitis On study for 122 weeks and then continued off-study for 90 weeks cGVHD stable/improved, thus weaned off therapy 
P13 Immune deficiency (ITK deficiency) Skin
Gut 
Steroid
Tacrolimus
Vedolizumab 
  • Acute GVHD: skin

  • Liver transplant before HCT for genetic liver failure; complicated by wound dehiscence and intra-abdominal infection requiring abdominal wash out and biliary duct dilation requiring ERCP and stent placement in the common bile duct.

  • Poor graft function despite stem cell boost

  • Persistent HHV-6 viremia

  • Chronic pericardial effusion

 
4.1 Overall CR
Skin CR
Gut CR 
Bacteremia Given persistent graft failure, infections, and multiorgan dysfunction, switched to comfort care 
P14 B-ALL Skin
Liver
Oral 
Steroid
MMF 
  • Acute GVHD: skin

 
10.4 Overall PR
Skin SD
Liver PR 
 Mild improvement in skin but early plateau; added ruxolitinib & stopped IL-2 
P15 Myeloproliferative disorder (JAK2 mutation) Skin
Liver
Ocular 
Steroid
Ruxolitinib 
  • Acute GVHD status unknown

  • Bilateral breast tissue ulceration/sclerodermatous change with MDR PsA that preceded starting IL-2

 
15.9 Overall PR
Skin PR
Liver CR 
Recurrence of breast tissue infection Malaise at daily dosing, switched to every other day
Discontinued, because unavailable in home country 

AKI, acute kidney injury; ALD, adrenoleukodystrophy; AML, acute myeloid leukemia; B-ALL, B-cell acute lymphoblastic leukemia; BK, BK virus; CMV, cytomegalovirus; CVL, central venous line; EBV, Epstein-Barr virus; ERCP, endoscopic retrograde cholangiopancreatography; fHLH, familial hemophagocytic lymphohistiocytosis; HHV-6, human herpesvirus 6; ITK, interleukin-2-inducible T-cell kinase; JMF, joints, muscles, fascia; MDR, multidrug resistant; MMF, mycophenolate mofetil; SAA, severe aplastic anemia; PsA, Pseudomonas; WAS, Wiskott-Aldrich syndrome.

Indicates had been stopped before starting IL-2; otherwise, therapies active at time of IL-2 start.

Developed lung involvement after start of IL-2 in the setting of aspergillosis.

Hypopigmentation, vitiligo; not part of official NIH scoring therefore not used for response (although improved).

LD IL-2 safety and tolerability

The median duration on therapy for our cohort was 462 days (range, 8-1489 days). Most patients received 1 × 106 IU/m2 per day, which was the previously identified maximum tolerated dose.20 However, 3 patients (P2, P9, and P15) requested a lowering of the dose because of discomfort or malaise associated with the daily subcutaneous injections (decreased to 0.33 × 106 IU/m2 per day for P2; 1 × 106 IU/m2 every other day for P9 and P15), which subsequently improved. P8 had dose reduction to 50% in the setting of reduced renal function. Most dose reductions occurred within the first 2 months of therapy.

No patient with a transplant indication of malignancy experienced relapse on LD IL-2 nor were there any serious adverse events directly attributable to LD IL-2 therapy. However, there were several adverse events that occurred during IL-2 therapy. P4 had a flare of preexisting alloimmune-mediated chronic relapsing encephalomyelitis upon steroid wean. P12 had a recurrence of pancreatitis after restarting steroids, and P15 had a recurrence of a multidrug resistant Pseudomonas breast abscess. In addition, 2 patients developed new infections, which occurred in the setting of multiple immune suppressive agents and cannot be attributed to LD IL-2 alone. P2 and P3 developed pulmonary aspergillosis during LD IL-2 therapy while also receiving 10 mg twice daily of ruxolitinib.

Clinical response and survival outcomes

Thirteen of 15 patients were evaluable for clinical response based on duration of therapy of >4 weeks. Two patients (P8 and P11) received <4 weeks of therapy (27 and 8 days, respectively) and were not considered evaluable for response. The ORR was 85% (11 of 13), comprised of 5 CR and 6 PR. The median time required to achieve PR in any involved organ was 8.3 weeks (range, 4-69 weeks) (Figure 1). There were 4 organ-specific PR by 6 weeks (2, skin; 1, liver; 1, joints-muscle-fascia). The median time to CR in any involved organ was 27 weeks (range, 8-80 weeks). Two patients (P7 and P10) had stable disease (SD). P10 had improvement of alopecia and vitiligo from cGVHD but these features are not included in the NIH skin scoring system.

Figure 1.

Swimmer plot outlining the timeline for clinical response in each patient’s best-response organ. These courses may be cross referenced against details in Table 2, which shows cotherapies and active organs at the time of LD IL-2 start (time 0 on x-axis). CsA, cyclosporine; IPA, invasive pulmonary aspergillosis; ITP, immune thrombocytopenia; MMF, mycophenolate mofetil; MOF, multiorgan failure; PsA, Pseudomonas; PNA, pneumonia; Tacro, tacrolimus; Vedo, vedolizumab.

Figure 1.

Swimmer plot outlining the timeline for clinical response in each patient’s best-response organ. These courses may be cross referenced against details in Table 2, which shows cotherapies and active organs at the time of LD IL-2 start (time 0 on x-axis). CsA, cyclosporine; IPA, invasive pulmonary aspergillosis; ITP, immune thrombocytopenia; MMF, mycophenolate mofetil; MOF, multiorgan failure; PsA, Pseudomonas; PNA, pneumonia; Tacro, tacrolimus; Vedo, vedolizumab.

Close modal

Patients P7 and P12 were initially enrolled in the pediatric cohort of the phase 1 clinical trial (NCT02318082) of intrapatient dose-escalated LD IL-2.18 P7, with isolated lung involvement, had a PR in the lungs at week 8 on study and continued extended-duration therapy for another 89 weeks before discontinuing because of progressive disease in the setting of a pulmonary infection. However, his lung cGVHD progressed even more rapidly when IL-2 was briefly discontinued. Thus, he was restarted on LD IL-2 off study, 9 weeks later, and was able to maintain SD over the next 2 years. P12 received 122 weeks of therapy on the same phase 1 clinical trial but had to discontinue on study because of shortage of study drug supply. However, he was able to obtain insurance coverage for aldesleukin and continued receiving LD IL-2 off study for another 90 weeks without interruption. The follow-up time in the prior publication was not long enough to capture his CR for lung at 80 weeks of therapy, and we include his full course to demonstrate safety for a total of 212 weeks of therapy.18 

Several patients with multiorgan involvement with an overall PR had organ-specific CR. Figure 2A-B show the organ-specific response rate in our cohort. The highest rates of CR were seen for liver (4 of 5 patients) and skin GVHD (5 of 7 patients), the latter of which reflects improvements in both erythroderma/desquamation as well as scleroderma (Figure 2C and D, respectively). In the 6 patients with lung involvement, there was 1 CR, 1 PR, and 4 SD based on formal NIH response criteria. However, several of the patients with SD showed improvement of percentage predicted forced expiratory volume in the first second (%FEV1) by pulmonary function testing despite not meeting objective PR criteria (supplemental Figure 1). For example, P2 initially had improvement in %FEV1 from 46% at baseline to 54% at 36 weeks on therapy, although she subsequently had worsening back down to 43%, 14 weeks later, after a presumed pulmonary aspergillosis infection. P3 developed new-onset bronchiolitis obliterans during his first course of LD IL-2 after pulmonary aspergillosis but then had improvement in %FEV1 from 30% at the start of his second course of LD IL-2 to 37% after 37 weeks of therapy. The CR shown for lung in P12 reflects pulmonary GVHD considered to be mixed cryptogenic organizing pneumonia/bronchiolitis obliterans syndrome. Of note, P12 also achieved CR of his liver and skin cGVHD.

Figure 2.

Clinical responses to LD IL-2 therapy. (A) Heatmap showing individual clinical responses for patients on a per-organ basis. (B) Summary of organ-specific clinical responses to LD IL-2 therapy. ORR is shown as percentage above bars. Number of patients with individual response (count) are embedded within each bar. (C) Improvement in erythroderma and desquamation after 6 weeks on therapy. (D) Improvement in scleroderma and hyperpigmentation at 14 weeks on therapy. ∗P3 initially started LD IL-2 for liver involvement and subsequently developed pulmonary cGVHD in the setting of pulmonary aspergillosis.

Figure 2.

Clinical responses to LD IL-2 therapy. (A) Heatmap showing individual clinical responses for patients on a per-organ basis. (B) Summary of organ-specific clinical responses to LD IL-2 therapy. ORR is shown as percentage above bars. Number of patients with individual response (count) are embedded within each bar. (C) Improvement in erythroderma and desquamation after 6 weeks on therapy. (D) Improvement in scleroderma and hyperpigmentation at 14 weeks on therapy. ∗P3 initially started LD IL-2 for liver involvement and subsequently developed pulmonary cGVHD in the setting of pulmonary aspergillosis.

Close modal

In addition to improvement in clinical features, many patients were able to significantly wean corticosteroid dose while receiving LD IL-2 (Figure 3A-B). All of the patients who were evaluable (n = 13) were on corticosteroids at LD IL-2 start with a median steroid dose of 0.79 mg/kg per day (range, 0.13-1 mg/kg per day). Most providers chose to wait until at least 4 weeks on LD IL-2 therapy before attempting a steroid wean. By week 8, the median dose was 0.50 mg/kg per day (range, 0.07-1 mg/kg per day) and median reduction from baseline was 30% (range, −72% to 0%) (P = 0.002). The median dose at IL-2 discontinuation was 0.09 mg/kg per day (range, 0-0.94 mg/kg per day) and median reduction from baseline was 56% (range, −100% to 30.8%) (P = 0.003) (supplemental Table 1). Six patients were able to permanently discontinue corticosteroid therapy after 16 to 20 weeks on LD IL-2.

Figure 3.

Steroid wean during LD IL-2 course. (A) Waterfall plots show decrease in steroid dose over time as compared with the dose at time of LD IL-2 start. (B) Steroid dose in mg/kg per day while on LD IL-2 therapy shown as mean with standard deviation in patients who were evaluable (n = 13). Baseline refers to steroid dose at time of LD IL-2 initiation. Final refers to steroid dose at time of LD IL-2 discontinuation. ∗∗P < .01.

Figure 3.

Steroid wean during LD IL-2 course. (A) Waterfall plots show decrease in steroid dose over time as compared with the dose at time of LD IL-2 start. (B) Steroid dose in mg/kg per day while on LD IL-2 therapy shown as mean with standard deviation in patients who were evaluable (n = 13). Baseline refers to steroid dose at time of LD IL-2 initiation. Final refers to steroid dose at time of LD IL-2 discontinuation. ∗∗P < .01.

Close modal

There were 2 deaths among the entire cohort of 15 patients with a median follow-up time among survivors of 22 months (range, 6-61 months). One-year survival was 86% (95% confidence interval, 53-96). Patient P8 was in liver failure from refractory liver cGVHD at the time of LD IL-2 initiation and also had ocular and oral involvement. He had no improvement of his ocular or oral cGVHD and died within 1 month of starting IL-2 therapy because of progressive liver failure. Patient P13 had a CR to LD IL-2 for skin and gut cGVHD but subsequently died from infection due to prolonged neutropenia in the setting of persistent human herpesvirus 6 viremia and poor graft function.

Clinical laboratory parameters

Although transient thrombocytopenia has been reported in previous studies of LD IL-2, IL-2–related cytopenias were not observed in this cohort. P11 stopped therapy after 8 days because of grade 4 thrombocytopenia but this was later determined to be from immune thrombocytopenia. As observed in prior LD IL-2 trials, eosinophilia, defined as an absolute eosinophil count of >1000/μL, was seen in several patients (supplemental Figure 2A). There were no clear clinical consequences of the eosinophilia, and there were no associated features of a systemic drug reaction. Eosinophilia resolved upon IL-2 discontinuation (supplemental Figure 2B).

Treg expansion

Because patients were not on a clinical trial, Treg numbers and other immune cell subsets were not measured consistently. However, lymphocyte subset data, performed over time in the clinical laboratory at Boston Children’s Hospital, were available in the medical record for 9 patients (Figure 4). As expected, there was preferential expansion of Tregs over conventional T cells (Tcons). The median Treg:Tcon ratio was 0.12 (range, 0.07-0.30) at baseline and increased to 0.52 (range, 0.28-1.26) at week 8 to 12 (P = 0.0039) (supplemental Table 2). The median peak fold increase in the Treg:Tcon ratio over baseline was 2.8 (range, 2-19.8), which occurred by 8 weeks on therapy (supplemental Table 3). Interestingly, patients receiving concurrent ruxolitinib (n = 3 with lymphocyte subsets) and calcineurin inhibitors (n = 3 with lymphocyte subsets) showed similar Treg expansion to patients not on these therapies (supplemental Table 3). Similarly, there was no clear impact of the initial steroid dose on Treg expansion (P5 and P10 started LD IL-2 while on 1mg/kg per day steroids and had peak Treg:Tcon fold expansions of 19.8 and 2.8, respectively). The Treg:Tcon ratio increased in all patients on LD IL-2 therapy (supplemental Figure 3) but numbers were insufficient to assess correlation with clinical response. NK cells also express the high-affinity IL-2 receptor and expand in response to LD IL-2.21 In this cohort, the NK cell population showed expected expansion on therapy with a median cell count per microliter at baseline of 134 (range, 25-507 cells per μL) compared with 424 cells per μL (range, 106.5-1794 cells per μL) at week 8 to 12 (P = 0.012; supplemental Table 2). The CD8+ T-cell fraction also increased from a median cell count per microliter at baseline of 156 (range, 58-694 cells per μL) compared with 378 (range, 31-1390 cells per μL) at week 8 to 12 (P = 0.027). There was no significant increase in B-cell count by week 8 to 12.

Figure 4.

Immune cell responses to LD IL-2 therapy. Absolute cell counts for lymphocyte subsets including CD4+ Tregs, CD4+ Tcons, CD8+ T cells, NK cells, and B cells as well as Treg:Tcon ratio are shown at baseline and after 8 to 12 weeks of LD IL-2 therapy. If patients had both week 8 (W8) and week 12 (W12) values, these were averaged. Baseline refers to the value at the measurement directly before LD IL-2 was initiated. ∗P < .05, ∗∗P < .01, ∗∗∗P < .001.

Figure 4.

Immune cell responses to LD IL-2 therapy. Absolute cell counts for lymphocyte subsets including CD4+ Tregs, CD4+ Tcons, CD8+ T cells, NK cells, and B cells as well as Treg:Tcon ratio are shown at baseline and after 8 to 12 weeks of LD IL-2 therapy. If patients had both week 8 (W8) and week 12 (W12) values, these were averaged. Baseline refers to the value at the measurement directly before LD IL-2 was initiated. ∗P < .05, ∗∗P < .01, ∗∗∗P < .001.

Close modal

LD IL-2 is a safe and well-tolerated therapy with promising efficacy for cGVHD and autoimmune disease.13-18,22-25 Although FDA-approved at high doses for renal cell carcinoma and metastatic melanoma since the 1990s, recombinant IL-2 is not yet approved at low doses for the promotion of immune tolerance in auto- and allo-immunity. Thus, use for these indications, to date, has been restricted to the clinical trial setting, almost exclusively in adult patients. Although clinical trials are well-controlled settings that allow for standardized assessment and correlative studies, they are less permissive to the practical aspects of therapy such as dose adjustments, changing concomitant therapies, and intermittent disruptions of therapy. Our study reports the first off-study clinical experience using LD IL-2. We also build on our center’s unique experience using LD IL-2 in pediatric patients with refractory cGVHD, including patients as young as 1.2 years of age.

As with our previous pediatric trial experience, we found that LD IL-2 is well tolerated, with the majority of patients continuing daily subcutaneous injections for >1 year. In addition, we observed a high ORR in pediatric patients with cGVHD that had failed multiple lines of prior therapy. All patients were started on our previously determined maximum tolerated dose of 1 × 106 IU/m2 per day, which was sustained daily for the entire duration of therapy with a few exceptions. Three patients electively reduced their dose because of malaise or discomfort. Despite these alternate dosing regimens, the ORR in our real-world study was 85%, which is similar to the response rate observed in our previous phase 1 dose-escalation study in children (ORR, 82%; n = 11).18 Furthermore, the 2 nonresponders in this off-study cohort were not patients for whom LD IL-2 doses had been reduced. As with this previous study, PR was seen in most patients by 8 weeks on therapy, and several patients attained organ-specific CR with extended-duration therapy (ie, >8 weeks), particularly for cutaneous and liver manifestations of cGVHD. Although steroid weaning practices are not standardized and do not always reflect improvement of cGVHD, almost all patients in this cohort were able to reduce their steroid dose without experiencing a cGVHD flare, including 6 patients who were able to discontinue altogether. Limiting steroid exposure is important for all patients, but it is particularly important in children to permit healthy growth, development, and metabolism. Overall, we recapitulated our previous on-study response rate despite dose reduction or alternate day dosing regimens for several patients. Thus, regimens other than daily dosing for LD IL-2 may also be efficacious.

Different dosing regimens of LD IL-2 have also been used for indications other than cGVHD. Our daily dosing regimen, while still relatively “low dose,” is notably higher compared with another published report of LD IL-2 in a phase 1/2 trial for pediatric patients with type I diabetes.22 In that study, IL-2 was given for 5 days and then once every 2 weeks for a year, with the highest dose being 0.5 × 106 IU/m2. There were no serious adverse events, and there was a dose-dependent correlation with Treg expansion, with “Treg-high” responders having more preserved insulin production at 1 year. This strategy of more frequent dosing during an induction period followed by less frequent maintenance dosing has also been used in adult trials for systemic lupus erythematosus, which have shown that it is clinically well tolerated, expands the Treg fraction, and has early suggestions of efficacy.24,25 Larger studies with pharmacokinetic analysis of serum IL-2 levels will be needed to determine the optimal dosing regimen.

As expected, LD IL-2 preferentially expanded the Treg fraction resulting in a higher Treg:Tcon ratio in the subset of patients with available immune cell values in the medical record. Beyond increasing the absolute number of Tregs and the Treg:Tcon ratio, LD IL-2 is also thought to have a beneficial effect on Treg diversity and function.16,18 Previous work at our center demonstrated that LD IL-2 increases Treg T-cell receptor β clonotype diversity and that this corresponds to better clinical response. In a study of LD IL-2 for systemic lupus erythematosus, the expression of 55 different genes was upregulated in Tregs after 2 months on treatment when compared with baseline.26 These differentially expressed genes were associated with cellular activation and tissue homing, including an HLA-DR+ subset of Tregs showing homing toward the skin based on biopsy data.26 Although similar work has not been performed in cGVHD, this subpopulation of cells could explain why LD IL-2 appears to be particularly effective for skin cGVHD, including reversal of sclerotic skin as observed in 2 patients in our off-study cohort.

Lastly, because patients could be on multiple concomitant therapies, we explored whether cotherapies might impair Treg expansion from LD IL-2. This was not seen, as high starting steroid dose (median 0.79 mg/kg per day) or the presence of ruxolitinib or calcineurin inhibitors did not appear to impair Treg expansion (or clinical response for those without laboratory data). Ruxolitinib is of interest because the IL-2 receptor is a JAK 1/3 receptor. Although ruxolitinib is more specific for JAK 1/2-based cytokine receptors, there could theoretically be nonspecific blockade that could dampen the therapeutic effect of LD IL-2.27 This was not reflected in our small cohort, as 5 patients were concurrently receiving ruxolitinib at time of LD IL-2 start and 4 had objective clinical responses (1 inevaluable) with Treg expansion in all 3 patients with available immune cell data. Calcineurin inhibitors block IL-2 production by effector T cells, which may be detrimental to Tregs by eliminating their critical source of IL-2. However, by providing the IL-2 exogenously, this problem is overcome, and Tregs can still expand while on concurrent LD IL-2 therapy and calcineurin inhibition.28 The ability for LD IL-2 to pair well with other drugs makes it attractive for clinical use, although our findings require further validation with higher patient numbers.

Although we are encouraged by our collective pediatric data, which now includes 24 pediatric patients with multiple years of follow-up, it has become increasingly difficult to obtain insurance approval for LD IL-2 because new small molecules have achieved FDA approval for second-line therapy of cGVHD. While our results reflect only single-center experience, LD IL-2 has consistently been shown as a safe and effective therapy with 105 adult and 11 pediatric patients29 treated across 5 clinical trials with efficacy at least on par with the FDA-approved agents. It is our hope that a Children’s Oncology Group–sponsored trial of LD IL-2 for refractory cGVHD in children, currently in development (Carrie Kitko, personal communication, 30 March 2023), can help to establish efficacy in a multicenter setting and subsequently advance the approval status of LD IL-2 for this indication.

Additional work with larger patient numbers is needed to determine baseline predictive factors for response to IL-2 and mechanisms of clinical improvement in different organs. Currently, young age and cutaneous or liver cGVHD involvement appear to be reasonable clinical predictors (the latter 2 for an organ-specific response) in pediatric patients, which may differ from the adult experience in which there was a lower cutaneous response.29 This may be because of underlying age-related differences in thymic output and cGVHD biology. Our center has also shown that combination therapy of LD IL-2 with ex vivo expanded Tregs can promote persistence of adoptively transferred Tregs in a nonhuman primate model, which is serving as the impetus for an upcoming clinical trial.30 An advantage of an exogenous Treg therapy is that it is possible to engineer even greater IL-2 specificity by using orthogonal IL-2/IL-2 receptor pairs.31,32 However, even for IL-2 biologic monotherapy, efforts are being made to engineer IL-2 muteins that have a longer half-life while being selective for the high affinity IL-2 receptor.33,34 Although we have not yet identified any clinical adverse effects from expansion of non-Treg cell populations, further enhancing specificity might limit epiphenomena such as the eosinophilia observed in many patients on LD IL-2, which is thought to be due to the release of IL-5 from IL-2 exposed innate lymphoid cells.35 

In summary, daily subcutaneous LD IL-2 is safe and well tolerated in children as young as 1.2 years of age for years on therapy. The majority of children have at least a PR, which is generally detectable by 8 weeks. Responses for liver and skin cGVHD are particularly notable, with most patients achieving CR in these organs. Our study thus supports the use of LD IL-2 in SR-GVHD, which has a high clinical unmet need. In addition, these data make a compelling case for further exploring the use of LD IL-2 to promote immune tolerance in pediatric patients with autoimmune disease or after solid organ transplantation.

NIAID T32AI007512-36 (H.W.); ASH RTAF award (F.A.C.)

Contribution: H.W., M.K., L.S.K., J.R., J.K., and J.S.W. conceived and designed the study; F.A.C., S.B., C.D., S.F., L.G., J.H., L.E.L., H.L., and M.S. referred/recruited patients; H.W., M.K., J.H., and J.S.W. collected and assembled the data; H.T.K. analyzed and interpreted the data, and performed statistical analysis; and all authors wrote the manuscript.

Conflict-of-interest disclosure: J.H. serves on the scientific advisory board for Ella Ola. S.B.’s spouse is an employee of Takeda Pharmaceuticals. L.S.K. is on the scientific advisory board for HiFiBio; reports research funding from Kymab Limited, Magenta Therapeutics, bluebird bio, and Regeneron Pharmaceuticals; reports consulting fees from Equillium, FortySeven Inc, Novartis Inc, EMD Serono, Gilead Sciences, and Takeda Pharmaceuticals; and reports grants and personal fees from Bristol Myers Squibb. J.K. receives research support from BMS, Miltenyi Biotec, Novartis, Clinigen, Regeneron; serves on scientific advisory boards for Therakos, Cugene, and Biolojic Design; consults for Amgen, Gentibio, Equillium, EMD Serono/Merck, and Moderna. J.R. receives research funding from Amgen, Equillium, Kite/Gilead, and Novartis; serves on data safety monitoring committees for AvroBio; and serves on scientific advisory boards for Akron Biotech, Clade Therapeutics, Garuda Therapeutics, Immunitas Therapeutics, LifeVault Bio, Novartis, Rheos Medicines, Talaris Therapeutics, and TScan Therapeutics. J.S.W. is a full-time employee of Vor Biopharma. The remaining authors declare no competing financial interests.

Correspondence: Jennifer S. Whangbo, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Dana 3, 450 Brookline Ave, Boston, MA 02215; e-mail: jennifer_whangbo@dfci.harvard.edu.

1.
Baird
K
,
Cooke
K
,
Schultz
KR
.
Chronic graft-versus-host disease (GVHD) in children
.
Pediatr Clin North Am
.
2010
. ;
57
(
1
):
297
-
322
.
2.
Pidala
J
,
Kurland
B
,
Chai
X
, et al
.
Patient-reported quality of life is associated with severity of chronic graft-versus-host disease as measured by NIH criteria: report on baseline data from the Chronic GVHD Consortium
.
Blood
.
2011
. ;
117
(
17
):
4651
-
4657
.
3.
Zeiser
R
,
Blazar
BR
.
Pathophysiology of chronic graft-versus-host disease and therapeutic targets
.
N Engl J Med
.
2017
. ;
377
(
26
):
2565
-
2579
.
4.
Alho
AC
,
Kim
HT
,
Chammas
MJ
, et al
.
Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD
.
Blood
.
2016
. ;
127
(
5
):
646
-
657
.
5.
Kawano
Y
,
Kim
HT
,
Matsuoka
KI
, et al
.
Low telomerase activity in CD4+ regulatory T cells in patients with severe chronic GVHD after hematopoietic stem cell transplantation
.
Blood
.
2011
. ;
118
(
18
):
5021
-
5030
.
6.
Zorn
E
,
Kim
HT
,
Lee
SJ
, et al
.
Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease
.
Blood
.
2005
. ;
106
(
8
):
2903
-
2911
.
7.
Matsuoka
KI
,
Kim
HT
,
McDonough
S
, et al
.
Altered regulatory T cell homeostasis in patients with CD4+ lymphopenia following allogeneic hematopoietic stem cell transplantation
.
J Clin Invest
.
2010
. ;
120
(
5
):
1479
-
1493
.
8.
Toubai
T
,
Magenau
J
.
Immunopathology and biology-based treatment of steroid-refractory graft-versus-host disease
.
Blood
.
2020
. ;
136
(
4
):
429
-
440
.
9.
Wolff
D
,
Fatobene
G
,
Rocha
V
,
Kröger
N
,
Flowers
ME
.
Steroid-refractory chronic graft-versus-host disease: treatment options and patient management
.
Bone Marrow Transplant
.
2021
. ;
56
(
9
):
2079
-
2087
.
10.
Zeiser
R
,
Lee
SJ
.
Three US Food and Drug Administration–approved therapies for chronic GVHD
.
Blood
.
2022
. ;
139
(
11
):
1642
-
1645
.
11.
Cutler
CS
,
Koreth
J
,
Ritz
J
.
Mechanistic approaches for the prevention and treatment of chronic GVHD
.
Blood
.
2017
. ;
129
(
1
):
22
-
29
.
12.
Yu
A
,
Zhu
L
,
Altman
NH
,
Malek
TR
.
A low interleukin-2 receptor signaling threshold supports the development and homeostasis of T regulatory cells
.
Immunity
.
2009
. ;
30
(
2
):
204
-
217
.
13.
Matsuoka
K-I
,
Koreth
J
,
Kim
HT
, et al
.
Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease
.
Sci Transl Med
.
2013
. ;
5
(
179
):
179ra43
.
14.
Whangbo
JS
,
Kim
HT
,
Nikiforow
S
, et al
.
Functional analysis of clinical response to low-dose IL-2 in patients with refractory chronic graft-versus-host disease
.
Blood Adv
.
2019
. ;
3
(
7
):
984
-
994
.
15.
Hirakawa
M
,
Matos
T
,
Liu
H
, et al
.
Low-dose IL-2 selectively activates subsets of CD4+ Tregs and NK cells
.
JCI Insight
.
2016
. ;
1
(
18
):
e89278
.
16.
Koreth
J
,
Kim
HT
,
Jones
KT
, et al
.
Efficacy, durability, and response predictors of low-dose interleukin-2 therapy for chronic graft-versus-host disease
.
Blood
.
2016
. ;
128
(
1
):
130
-
137
.
17.
Belizaire
R
,
Kim
HT
,
Poryanda
SJ
, et al
.
Efficacy and immunologic effects of extracorporeal photopheresis plus interleukin-2 in chronic graft-versus-host disease
.
Blood Adv
.
2019
. ;
3
(
7
):
969
-
979
.
18.
Whangbo
JS
,
Kim
HT
,
Mirkovic
N
, et al
.
Dose-escalated interleukin-2 therapy for refractory chronic graft-versus-host disease in adults and children
.
Blood Adv
.
2019
. ;
3
(
17
):
2550
-
2561
.
19.
Jagasia
MH
,
Greinix
HT
,
Arora
M
, et al
.
National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. The 2014 diagnosis and staging working group report
.
Biol Blood Marrow Transplant
.
2015
. ;
21
(
3
):
389
-
401.e1
.
20.
Lee
SJ
,
Wolff
D
,
Kitko
C
, et al
.
Measuring therapeutic response in chronic graft-versus-host disease. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: IV. The 2014 response criteria working group report
.
Biol Blood Marrow Transplant
.
2015
. ;
21
(
6
):
984
-
999
.
21.
Koreth
J
,
Matsuoka
K
,
Kim
HT
, et al
.
Interleukin-2 and regulatory T cells in graft-vs-host disease
.
N Eng J Med
.
2011
. ;
365
(
22
):
2055
-
2066
.
22.
Rosenzwajg
M
,
Salet
R
,
Lorenzon
R
, et al
.
Low-dose IL-2 in children with recently diagnosed type 1 diabetes: a phase I/II randomised, double-blind, placebo-controlled, dose-finding study
.
Diabetologia
.
2020
. ;
63
(
9
):
1808
-
1821
.
23.
Rosenzwajg
M
,
Lorenzon
R
,
Cacoub
P
, et al
.
Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a single, open clinical trial
.
Ann Rheum Dis
.
2019
. ;
78
(
2
):
209
-
217
.
24.
Humrich
JY
,
Cacoub
P
,
Rosenzwajg
M
, et al
.
Low dose interleukin-2 therapy in active systemic lupus erythematosus (LUPIL-2): a multicentre, double-blind, randomised and placebo-controlled phase II trial
.
Ann Rheum Dis
.
2022
. ;
81
(
12
):
1685
-
1694
.
25.
He
J
,
Zhang
R
,
Shao
M
, et al
.
Efficacy and safety of low-dose IL-2 in the treatment of systemic lupus erythematosus: a randomised, double-blind, placebo-controlled trial
.
Ann Rheum Dis
.
2020
. ;
79
(
1
):
141
-
149
.
26.
Raeber
ME
,
Caspar
D
,
Zurbuchen
Y
, et al
.
Interleukin-2 immunotherapy reveals human regulatory T cell subsets with distinct functional and gatekeeper features
.
medRxiv
.
2022
. . Preprint posted online 15 November.
27.
Parampalli Yajnanarayana
S
,
Stübig
T
,
Cornez
I
, et al
.
JAK1/2 inhibition impairs T cell function in vitro and in patients with myeloproliferative neoplasms
.
Br J Haematol
.
2015
. ;
169
(
6
):
824
-
833
.
28.
Furukawa
A
,
Wisel
SA
,
Tang
Q
.
Impact of immune-modulatory drugs on regulatory T cell
.
Transplantation
.
2016
. ;
100
(
11
):
2288
-
2300
.
29.
Kim
HT
,
Koreth
J
,
Whangbo
J
, et al
.
Organ-specific response after low-dose interleukin-2 therapy for steroid-refractory chronic graft-versus-host-disease
.
Blood Adv
.
2022
. ;
6
(
15
):
4392
-
4402
.
30.
Furlan
SN
,
Singh
K
,
Lopez
C
, et al
.
IL-2 enhances ex vivo-expanded regulatory T-cell persistence after adoptive transfer
.
Blood Adv
.
2020
. ;
4
(
8
):
1594
-
1605
.
31.
Sockolosky
JT
,
Trotta
E
,
Parisi
G
, et al
.
Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes
.
Science
.
2018
. ;
359
(
6379
):
1037
-
1042
.
32.
Hirai
T
,
Ramos
TL
,
Lin
PY
, et al
.
Selective expansion of regulatory T cells using an orthogonal IL-2/IL-2 receptor system facilitates transplantation tolerance
.
J Clin Invest
.
2021
. ;
131
(
8
):
e139991
.
33.
de Picciotto
S
,
DeVita
N
,
Hsiao
CJ
, et al
.
Selective activation and expansion of regulatory T cells using lipid encapsulated mRNA encoding a long-acting IL-2 mutein
.
Nat Commun
.
2022
. ;
13
(
1
):
3866
.
34.
Ward
NC
,
Yu
A
,
Moro
A
, et al
.
IL-2/CD25: a long-acting fusion protein that promotes immune tolerance by selectively targeting the IL-2 Receptor on regulatory T cells
.
J Immunol
.
2018
. ;
201
(
9
):
2579
-
2592
.
35.
Van Gool
F
,
Molofsky
AB
,
Morar
MM
, et al
.
Interleukin-5-producing group 2 innate lymphoid cells control eosinophilia induced by interleukin-2 therapy
.
Blood
.
2014
. ;
124
(
24
):
3572
-
3576
.

Author notes

Data are available on request from the corresponding author, Jennifer S. Whangbo (jennifer_whangbo@dfci.harvard.edu).

The full-text version of this article contains a data supplement.

Supplemental data