Leflunomide Confers Rapid Recovery from COVID-19 and is Coupled with Temporal Immunologic Changes

Ada Alice Dona1,2#, James F Sanchez1#, Joycelynne M Palmer3, Timothy W. Synold4, Flavia Chiuppesi5, Sandra Thomas2, Enrico Caserta1,2, Mahmoud Singer1,2, Theophilus Tandoh1,2, Arnab Chowdhury3, Amrita Krishnan1, Michael Rosenzweig1, Don J Diamond5, Steven Rosen1*, Flavia Pichiorri1,2*, Sanjeet Dadwal6*

1Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA

2Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA

3Department of Computational and Quantitative Sciences, Beckman Research Institute, City of Hope, Duarte, CA

4Department of Cancer Biology, City of Hope, Duarte, CA

5Department of Experimental Therapeutics, City of Hope, Duarte, CA

6Department of Medicine, Division of Infectious Disease, City of Hope, Duarte, CA USA

#These authors contributed equally to this work.


Background: Vaccines for SARS-CoV-2 have been considerably effective in reducing rates of infection and severe COVID-19. However, many patients, especially those who are immunocompromised due to cancer or other factors, as well as individuals who are unable to receive vaccines or are in resource-poor countries, will continue to be at risk for COVID-19. We describe clinical, therapeutic, and immunologic correlatives in two patients with cancer and severe COVID-19 who were treated with leflunomide after failing to respond to standard-of-care comprising remdesivir and dexamethasone. Both patients had breast cancer and were on therapy for the malignancy.

Methods: The protocol is designed with the primary objective to assess the safety and tolerability of leflunomide in treating severe COVID-19 in patients with cancer. Leflunomide dosing consisted of a loading dose of 100 mg daily for the first three days, followed by daily dosing, at the assigned dose level (Dose Level 1: 40 mg, Dose Level -1, 20 mg; Dose Level 2, 60 mg), for an additional 11 days. At defined intervals, serial monitoring of blood samples for toxicity, pharmacokinetics, and immunologic correlative studies were performed, as well as nasopharyngeal swabs for PCR analysis of SARS-CoV-2.

Results: Preclinically, leflunomide impaired viral RNA replication, and clinically, it led to a rapid improvement in the two patients discussed herein. Both patients completely recovered, with minimal toxicities; all adverse events experienced were considered unrelated to leflunomide. Single-cell mass-cytometry analysis showed that leflunomide increased levels of CD8+ cytotoxic and terminal effector T cells and decreased naïve and memory B cells.

Conclusions: With ongoing COVID-19 transmission and occurrence of breakthrough infections in vaccinated individuals, including patients with cancer, therapeutic agents that target both the virus and host inflammatory response would be helpful despite the availability of currently approved anti-viral agents. Furthermore, from an access to care perspective, especially in resource-limited areas, an inexpensive, readily available, effective drug with existing safety data in humans is relevant in the real-world setting.


Background

Vaccines for SARS-CoV-2, the causative agent of COVID-19, have been considerably effective in reducing rates of infection and severe COVID-19. However, many individuals, including some patients with cancer, people who are unable to receive vaccines, or individuals in resource-poor countries, where access to antiviral medications, monoclonal antibodies, and vaccines is limited, will continue to be at risk for COVID-19. In infected patients, the non-specific innate immune response is followed by adaptive immunity mediated by both B cells and T cells,1 but patients with cancer and other disorders may be impacted by a dysregulated immune response.

Leflunomide is an FDA-approved, oral agent that has been commercially available since 1998. It is used for the treatment of rheumatoid arthritis as a single agent or in combination with methotrexate. Its primary mechanism of action is inhibiting de novo pyrimidine synthesis by targeting dihydroorotate dehydrogenase (DHODH)2, with an anti-proliferative effect in B- and T-lymphocytes3. Teriflunomide is the active primary metabolite of leflunomide.

Host-targeting antiviral drugs may be appealing by targeting the host machinery exploited by the virus, thereby potentially applying to a wide range of viruses and viral strains. To this end, repurposed drugs are especially suitable, as many of the extensive preclinical validation and toxicity studies have already been performed. As a DHODH inhibitor, leflunomide is worthy of study for SARS-CoV-2, an RNA virus, particularly because of the virus’s high content of uracil4, one of the two nucleobases inhibited by leflunomide. Leflunomide has been used in difficult-to-treat cytomegalovirus infections in patients undergoing hematopoietic cell transplantation and in polyoma virus (BK virus) hemorrhagic cystitis when no alternatives are available.

Methods

The protocol is designed as a phase 1/ 2 trial with the primary objective to assess the safety and tolerability of leflunomide in treating severe COVID-19 in patients with cancer (NCT04532372). The study was performed in accordance with the provisions of the Declaration of Helsinki and approved by the City of Hope Institutional Review Board. All participants gave written informed consent. Leflunomide dosing comprised a loading dose of 100 mg daily for the first three days, followed by daily dosing at the assigned dose level (Dose Level 1: 40 mg, Dose Level -1, 20 mg; Dose Level 2, 60 mg), for an additional 11 days. At defined intervals, serial monitoring of blood samples for toxicity, pharmacokinetics, and immunologic correlative studies were performed, as well as nasopharyngeal swabs for PCR analysis of SARS-CoV-2.

Primary Samples

Biospecimen collection and immunological analyses are detailed below. Briefly, peripheral blood from healthy donors was obtained from the institutional hematopoietic tissue repository. Specifically, the cellular fraction of the peripheral blood mononuclear cells (PBMCs) was isolated using Ficoll-Paque Plus (GE Healthcare) following the manufacturer’s instructions.

Mass Cytometry (CyTOF) Staining and Acquisition

A total of 2-4x106 PBMCs were stained with a panel containing 30 metal-conjugated antibodies (Sup. Table 1) according to Fluidigm's protocol for Maxpar Antibody Labeling (PRD002 Rev 12). PBMCs derived from the clinical trial were stained with Fluidigm's Maxpar Direct Immune Profiling Assay Cell Staining (PN 400286 B1). Samples were acquired, exported as FCS files, and normalized on Fluidigm's Helios (Software 7.0.5189).

Supplementary Table 1: Maxpar direct immune profiling assay 30-marker panel with clones and heavy metals (Fluidigm)

Target

Clone

Metal

Anti-human CD45

HI30

89Y

Live/dead 103Rh-Intercalator (500 μM)

N/A

103Rh

Anti-human CD196/CCR6

G034E3

141Pr

Anti-human CD123

6H6

143Nd

Anti-human CD19

HIB19

144Nd

Anti-human CD4

RPA-T4

145Nd

Anti-human CD8a

RPA-T8

146Nd

Anti-human CD11c

Bu15

147Sm

Anti-human CD16

3G8

148Nd

Anti-human CD45RO

UCHL1

149Sm

Anti-human CD45RA

HI100

150Nd

Anti-human CD161

HP-3G10

151Eu

Anti-human CD194/CCR4

L291H4

152Sm

Anti-human CD25

BC96

153Eu

Anti-human CD27

O323

154Sm

Anti-human CD57

HCD57

155Gd

Anti-human CD183/CXCR3

G025H7

156Gd

Anti-human CD185/CXCR5

J252D4

158Gd

Anti-human CD28

CD28.2

160Gd

Anti-human CD38

HB-7

161Dy

Anti-human CD56/NCAM

NCAM16.2

163Dy

Anti-human TCRgd

B1

164Dy

Anti-human CD294

BM16

166Er

Anti-human CD197/CCR7

G043H7

167Er

Anti-human CD14

63D3

168Er

Anti-human CD3

UCHT1

170Er

Anti-human CD20

2H7

171Yb

Anti-human CD66b

G10F5

172Yb

Anti-human HLA-DR

LN3

173Yb

Anti-human IgD

IA6-2

174Yb

Anti-human CD127

A019D5

176Yb

CyTOF Analysis

Non-custom panel analysis was analyzed using Maxpar Pathsetter™ software powered by GemStone 2.0.41, Verity Software House, Topsham, Maine (Version 2.0.45). The FCS files were also analyzed using FlowJo™ Software (Windows edition, Version 10.6. Becton Dickinson Company; 2019), and the Cytobank© platform (https://www.cytobank.org) (Cytobank, Inc., Mountain View, CA) was used for gating, tSNE plotting, and FlowSOM.

Pharmacokinetic Analysis

Pharmacokinetic sampling was performed per protocol during active treatment with leflunomide. Samples were collected prior to initiation of drug administration, daily for days +1 to +7 of treatment, and subsequently daily if the subject was hospitalized, or during visits if the subject was outpatient. Plasma was analyzed for total (bound and unbound) and free (unbound) teriflunomide concentrations using a previously described LC-MS/MS method5. Briefly, following addition of a deuterated teriflunomide internal standard, plasma (total drug) or ultrafiltered plasma (free drug) samples were prepared by protein precipitation using 3:1 ice cold methanol. The resulting protein-free plasma or ultrafiltered plasma was further diluted 10,000-fold with 0.5 mM ammonium acetate, 0.025% formic acid in 75% methanol, and 10 µl was injected for analysis. The lower limits of quantitation of the assay were 30 µg/ml and 30 ng/ml in plasma and ultrafiltered plasma, respectively.

DLT Definitions

Dose limiting toxicity (DLT) was defined as any of the following toxicities that were at least possibly related to leflunomide:

  • Hematologic DLT events (any adverse events [AEs] considered at least possibly attributable to study treatment):
  • For patients with solid tumor malignancies, all grade 3 or 4 hematologic AEs
  • For patients with hematologic malignancies, the following AEs would be considered DLTs if they were determined as not attributable to underlying hematological malignancy.
  • Grade 4 neutropenia (absolute neutrophil count [ANC] <500/mm3)
  • Grade 3 or 4 febrile neutropenia
  • Grade 4 thrombocytopenia (<25,000/mm3)
  • Grade 3 thrombocytopenia (<50,000/mm3) with bleeding
  • Non-Hematologic AEs (any treatment emergent AEs):
  • ≥ Grade 3 non-hematologic toxicity excepting the following:
  • Alopecia
  • Grade 3 nausea/vomiting/diarrhea for less than 72 hours treated with adequate antiemetic and other supportive care
  • Grade 3 fatigue for < 1 week
  • ≥ Grade 3 electrolyte abnormalities that are not clinically complicated and resolve spontaneously or to conventional medical interventions within 72 hours
  • ≥ Grade 3 amylase or lipase elevation not associated with symptoms or clinical manifestations of pancreatitis
  • Treatment-emergent increase in serum ALT or AST to > 3x ULN associated with an increase in serum total bilirubin to > 2x ULN (consistent with Hy’s Law).
  • Any treatment-emergent Grade 5 AE that occurs during the 28-day treatment period that is not due to underlying malignancy would be considered a DLT.

Cholestyramine

Leflunomide is reabsorbed from the gastrointestinal tract and consequently is detectable in the body for up to 2 years. Cholestyramine, a bile acid sequestrant, fixes the metabolite, preventing reabsorption and expediting drug elimination. We administered cholestyramine daily starting on day 29 until plasma levels of teriflunomide were less than 0.02 mg/L by two separate measurements 14 days apart.

Cell culture

Multiple myeloma (MM) cell lines MM.1S and U266 were purchased from ATCC. All cell lines were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) (Cat. #019K8420, Sigma), 100 IU/mL penicillin, and 100 μg/mL streptomycin (Cat.#15140-122, Gibco).

Immunoblotting

Cells lyses in RIPA buffer (89901, Thermo Scientific) were supplemented with protease and phosphatase inhibitors, and then sonicated (30” Pulse ON 02” Pulse OFF 03” 60% amplification). Lysates were then clarified by spinning at 14,000 rpm at 4°C and protein concentration quantified by BCA Protein Assay (23227, Thermo Scientific). Forty micrograms of proteins were denatured in boiling SDS sample buffer, resolved on 4%-20% gradient gels (Cat.# 5671093, Bio-Rad), and transferred to nitrocellulose membranes (Cat.# 1704271 Bio-Rad). After blocking nonspecific binding of antibody with 5% BSA (Fisher BioReagents), blots were probed with anti-RV σ-NS (1:1000; provided by Oncolytics Biotech Inc.) to assess viral replication for σ1 protein, or GAPDH (Cat.# sc-32233, Santa Cruz Biotechnology) as internal control. Blots were washed three times for 15 minutes with TBST 1X and stained with horseradish peroxidase (HRP)-conjugated secondary antibodies (diluted 1:4000) for 2hrs at room temperature. Primary antibodies were detected by binding with secondary antibodies donkey anti-goat IgG (H+L) (Cat.# A16005, Invitrogen) and goat anti-mouse IgG-HRP (NA931, GE Healthcare), and using an enhanced chemiluminescent visualization system (Cat.#RPN2209 ECL Western Blotting Detection Reagents, GE Healthcare). Primary and secondary antibodies were diluted according to the manufacturer instructions. The bands were quantified by densitometry analyses using Image Lab program (Biorad) and normalized to GAPDH.

RNA isolation and analysis

Total cellular RNA was extracted by using TRIZOL reagent (Cat. #15596018 Invitrogen Corporation) and RNA Clean-Up and Concentration Kit (Cat. #43200 Norgen) according to the manufacturer’s protocols. cDNA synthesis was performed by using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Cat# 4368814). Reverse transcription reactions were run using a Mastercycler pro. Quantitative real time-PCR (qRT-PCR) was performed with the TaqMan method (Applied Biosystems), according to the manufacturer’s instructions. The appropriate TaqMan probes for mRNA quantification were purchased from Applied Biosystems, and all reactions were performed in triplicate. The following probes were used: (HS99999905_m1) GAPDH used as endogenous control; (HS00989291_m1) INF-γ; (Hs00961622 m1) IL-10; (Hs00174131 m1) IL-6; (Hs01077958 S1) IFNB1; (Hs00174128 m1) TNFalpha; (Hs00265051 S1) IFNA2.

For the quantification of viral RNA of genomes extracted from infected PBMCs from healthy donors and MM cell lines, q-RT-PCR reactions were conducted using the PowerUp SYBR Green Master Mix (Applied Biosystems Cat.# 4367659) according to the manufacturer’s instructions and the following primers: Reo9: 5′-TG CGC AAG AGG CAG CAA TCG-3′ and Reo10: 5′-TT CGC GGG CCT CGC ACA TTC-3′; GAPDH FD: 5′- CTG CAC CAC CAA CTG CTT -3′ and GAPDH RV: 5′- CAT GAC GGC AGG TCA GGT -3′.

Cytokine measurements

Samples were analyzed for 30 cytokines using the Human Cytokine Thirty-Plex Antibody Magnetic Bead Kit (Invitrogen, Camarillo, CA) as per the manufacturer’s protocol. Briefly, Invitrogen’s multiplex bead solution was vortexed for 30 seconds, and 25 µl was added to each well of a flat-bottom 96 well microplate. Samples (e.g., plasma) were diluted 1:2 with assay diluent and loaded into the wells containing 50 µl of incubation buffer. Cytokine standards were reconstituted with assay diluent, and serial dilutions of cytokine standards were prepared in parallel and added to the plate. Plates were incubated on a shaker at 500 rpm in the dark at room temperature for 2 hours. The plate was then applied to a magnetic capture device and washed three times with 200 µl of wash buffer. After the final wash, 200 µl of a biotinylated detection antibody mixture was added to each well, and the plate was incubated on a shaker for 1 hour. After washing again three times with 200 µl of wash buffer, streptavidin-phycoerythrin (100 µl) was added to the wells. The plate was incubated on a plate shaker for another 30 minutes and washed three times, after which the beads were resuspended in 150 µl of wash buffer and shaken for 1 minute. Finally, the assay plate was transferred to a Flexmap 3D Luminex system (Luminex Corp, Austin, TX) for analysis. Cytokine concentrations were calculated using Bio-Plex Manager 6.0 software with a five parameter curve-fitting algorithm applied for standard curve calculations for duplicate samples.

Results and Discussion

Previous data have demonstrated the preclinical anti-viral activity of DHODH inhibitors against RNA viruses such as influenza, Ebola, and Zika viruses, as well as SARS-CoV-26. In support of the importance of DHODH in viral replication, viral growth was largely inhibited in DHODH knockout cells, and addition of uracil and cytosine (the pyrimidine bases) restored viral activity6.

Reovirus serotype 3–dearing strain (RV) is a naturally occurring, ubiquitous, nonenveloped human reovirus with a genome that consists of 10 segments of double-stranded RNA. As with SARS-CoV-2, RV can infect human and animals and never passes through a DNA phase. It actively replicates its RNA genome and induces a strong anti-viral immune response. The two viruses primarily differ in the composition of the envelope and the fact that coronaviruses need to synthetize the RNA negative strand before initiating transcription. Our preliminary data show that leflunomide significantly arrested RV productive infection in cancer cells, as shown by decreased capsid formation and genome replication (Sup. Fig. 1A-F). Impairment in viral replication was also observed when PBMCs were treated with teriflunomide, the active metabolite of leflunomide, in combination with RV, compared to the virus alone (Sup. Fig. 1G-H). Our data also show that, in the ex vivo setting, the addition of teriflunomide to RV-infected PBMCs significantly decreased the expression of highly inflammatory cytokines such as IL-6 and GM-CSF (Sup. Fig. 1I-J) but concomitantly enhanced the anti-viral interferon I (IFN-α and IFN-β) response and TNF-α (Sup. Fig. 1K-L-M). The antiviral activity of leflunomide aligns with the experience of the drug as an agent against cytomegalovirus (CMV) and polyoma BK virus infections in immunocompromised hosts7,8.

JISS-22-1241-fig16

Supplementary Figure 1: A-B-C)Western Blot analysis of σ-NS viral protein in U266 cells after 6, 16 and 48hrs of treatment with teriflunomide (100 µm) and/or RV (5 MOI) infection (A), and q-RT-PCR for the viral genome expression after 16hrs (B) or 48hrs (C), normalized compared to control GAPDH and expressed as the mean ± SEM of triplicates in fold-change compared to the control; D-E-F) Western Blot analysis of σ-NS viral protein in MM1.S cells after 16 and 48hrs of treatment with teriflunomide (100 µm) and/or RV (5 MOI) infection (D), and q-RT-PCR for the viral genome expression after 16hrs (E) or 48hrs (F), normalized compared to control GAPDH and expressed as the mean ± SEM of triplicates in fold-change compared to the control; G-H) Western Blot analysis of (σ-NS) viral protein in healthy donor PBMC treated for 48hrs with teriflunomide (100 µM) and infected with RV (5 MOI); H) After 16hrs, RNA was isolated, and expression of the RV genome was determined by q-RT-PCR. Data were normalized compared to control GAPDH and expressed as the mean ± SEM of triplicates as fold-change compared to the control; I-J-K-L-M) PBMCs obtained from healthy donors were treated with teriflunomide (100 µM) and/or RV (5 MOI) and infected for 16hrs to assess the cytokine (IL-6, GM-CSF, IFN-α, IFN-β, TNF-α) mRNA expression profile by q-RT-PCR. For IL-6 and GM-CSF, data are expressed as the mean ± SEM (IL-6 and GM-CSF n=2 healthy donors; IFN-α, IFN-β and TNF-α n=3 healthy doors), normalized compared to control GAPDH; Comparisons among groups were performed by one-way ANOVA.

We furthermore describe our findings of two patients with breast cancer who were admitted to our COVID unit after worsening shortness of breath due to COVID-19. Both patients’ oxygen levels decreased to below 90%, requiring high-flow oxygen. Computed tomography (CT) of the chest revealed multifocal pneumonia in both patients (Fig. 1A-B). The patients began treatment with remdesivir, dexamethasone, cefepime, and azithromycin, with minimal improvement. No co-infections were identified, and the clinical condition was due to COVID-19.

We administered leflunomide to both patients following informed consent and confirmation of eligibility to participate in the clinical trial. After loading with leflunomide with a dose of 100 mg daily x 3 days, the patients received 40 mg daily for 11 additional days. Leflunomide is reabsorbed from the gastrointestinal tract and consequently is detectable in the body for up to 2 years. Cholestyramine, a bile acid sequestrant, fixes the metabolite, preventing reabsorption and expediting drug elimination. We administered cholestyramine daily starting on day 29 until plasma levels of teriflunomide were less than 0.02 mg/L by two separate measurements 14 days apart. Plasma concentrations of teriflunomide were also measured throughout the treatment period in both patients as previously described9. Maximum plasma concentrations (Cmax) achieved in the two subjects were 252.4 and 152.3 µmol/L, respectively, and the areas-under-the-curve (AUC) were 5598.0 and 3498.1 µmol/L x day. As shown in the concentration-versus-time plots (Sup. Fig. 2A), plasma concentrations were maintained above the in vitro EC50 against SARS-CoV-2 (26 µmol/L) 6 for >36 days.

JISS-22-1241-fig2

Figure 1: A-B)Serial CT chest scans from patient 1 (A) and 2 (B) with severe COVID-19 pneumonia showing peripheral multilobar ground glass opacities (red arrow) two days before the initiation of leflunomide treatment for patient 1 and Day 6 of treatment for patient 2. Scans obtained after 36 days (A) and after 93 days (B) on treatment show partial absorption of the abnormalities for both patients.

JISS-22-1241-fig3

Supplementary Figure 2: A)Teriflunomide plasma concentration-versus-time plots for 2 patients receiving leflunomide. The dashed line represents the in vitro EC50 for teriflunomide against SARS-CoV-2 (26 µmol/L). B-C) In-depth longitudinal immune profiling of two patients with COVID‐19 and breast cancer (A and B) treated with 40 mg leflunomide. A Maxpar Direct Immune Profiling System using a dry 30-marker antibody panel was employed. Hierarchical clustering and statistical mapping were performed algorithmically via the Cytobank© platform. vi-SNE analysis (iterations=1000, perplexity=100) are displayed in 2D plots using the resultant t-SNE 1 and t-SNE2 dimensions. High-fidelity FlowSOM (“self-organizing map”) (metacluster=10 and cluster=100) based on vi-SNE 2D plots showing 20 different immune-compartments was used; D) Line graphs showing B cell expression as a percentage of the total PBMC population in Patients 1 and 2 as measured by CyTOF; E) Line graphs showing CD8+ T cell expression as a percentage of the total PBMC population in Patients 1 and 2 as measured by CyTOF; F-G) t-SNE heatmaps showing overall CD8+ T cell expression in both patients.

JISS-22-1241-fig4

Supplementary Figure 2: H)Line graphs showing CD4+ T cell expression as a percentage of the total PBMC population in Patients 1 and 2 as measured by CyTOF; I-J) t-SNE heatmaps showing overall CD4+ T cell expression in both patients.

The patients had rapid improvement in symptoms and oxygenation and experienced a complete recovery as clearly shown from the CT scans of the lung before and after combination therapy that included leflunomide (Fig. 1A-B). The adverse events that were observed were minimal and considered unrelated to leflunomide. These findings are consistent with a pilot study in which 15 patients given leflunomide had improved viral clearance and hospital discharge rate when compared to 12 patients in a control arm10. Because leflunomide is an immune suppressive drug, which could potentially impair its anti-viral activity, we decided to follow the immune changes of these patients during and after the course of treatment. A 30-marker single cell mass cytometry (CyTOF) antibody panel (Sup. Table 1) designed to identify twenty different immune compartments (Sup. Table 2) was used to perform a longitudinal high dimensional immune profiling (Fig. 2A and Sup. Fig. 2B-C).

JISS-22-1244-fig5

Figure 2: A) In-depth longitudinal immune profiling of two patients with COVID‑19 and breast cancer treated with 40 mg leflunomide. A Maxpar Direct Immune Profiling System using a dry 30-marker antibody panel was employed. Hierarchical clustering and statistical mapping were performed algorithmically via the Cytobank© platform. vi-SNE analysis (iterations=1000, perplexity=100) are displayed in 2D plots using the resultant t-SNE 1 and t-SNE 2 dimensions. High-fidelity FlowSOM (“self-organizing map”) (metacluster=10 and cluster=100) based on vi-SNE 2D plots showing 20 different immune-compartments was used; B) Bar graphs representing the expression of naïve and memory B cell subpopulations through the course of treatment; C) t-SNE heatmap highlighting the increased expression of CD8+ cytotoxic T cells at baseline and on days 14 and 21 of treatment in both patients; D) Bar graphs showing effector memory and terminally differentiated effector T cells (TEMRA) T cell subsets in percentages compared to the total count PBMCs in both patients.

Supplementary Table 2: CyTOF gating strategy

Cell subsets

Model phenotypes

Naïve B-cells

CD45+ CD3- CD19+ CD20+ HLA-DR+ CD27-

Memory B-cells

CD45+ CD3- CD19+ CD20+ HLA-DR+ CD27+

Early NK

CD45+ CD3- CD19- CD14- CD56+ CD16-

Mature NK

CD45+ CD3- CD19- CD14- CD56+ CD16+

M-MDSCs

CD45+ CD3- CD19- CD11b+ CD33+ CD14+ HLA-DR-/dim

Classical Monos

CD45+ CD3- CD19- CD56- CD11b+ CD33+ HLA-DR+ CD14+ CD16-

Intermediate Monos

CD45+ CD3- CD19- CD56- CD11b+ CD33+ HLA-DR+ CD14+ CD16+

Non-Classical Monos

CD45+ CD3- CD19- CD56- CD11b+ CD33+ HLA-DR+ CD14- /dim CD16++

RV in Monos

CD45+ CD3- CD19- CD56- CD11b+ CD33+ HLA-DR+ CD14+ RV+

NKt-cells

CD45+ CD19- CD3+ CD56+

TCR γδ

CD45+ CD19- CD3+ CD56- TCRgd+

AE T-cells

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25+ CD127+

Memory Tregs

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25+ CD127low/- CD45RA-

Naïve Tregs

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25+ CD127low/- CD45RA+

CM CD8

CD45+ CD19- CD3+ CD56- TCRgd- CD8+ CD4- CD45RA- CCR7+ CD27+

EM CD8

CD45+ CD19- CD3+ CD56- TCRgd- CD8+ CD4- CD45RA- CCR7- CD27+

Naïve CD8

CD45+ CD19- CD3+ CD56- TCRgd- CD8+ CD4- CD45RA+ CCR7+ CD27+

TEMRA CD8

CD45+ CD19- CD3+ CD56- TCRgd- CD8+ CD4- CD45RA+ CCR7- CD27-

CM CD4

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25- CD127+CD45RA- CCR7+ CD27+

EM CD4

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25- CD127+CD45RA- CCR7- CD27+

Naïve CD4

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25- CD127+CD45RA+ CCR7+ CD27+

TEMRA CD4

CD45+ CD19- CD3+ CD56- TCRgd- CD8- CD4+ CD25- CD127+/dim CD45RA+ CCR7- CD27-

In both patients, we observed a substantial decrease in the total number of B cells following leflunomide treatment (Fig. 2B and Sup. Fig. 2D) (Sup. Tables 3 and 4). This finding is consistent with the regulatory activity of leflunomide on B cell proliferation that was previously observed in animals11. Although leflunomide has been considered an immunosuppressive drug by inhibiting cellular and humoral mediated responses12, in both patients, we observed a robust expansion in CD8+ cytotoxic T cells after 5 days from the initial dosing. This effect further increased upon subsequent doses, reaching maximum expansion at around 21 days after the initial treatment (Fig. 2C and Sup. Fig. 2E-G). Among the CD8+ cytotoxic T cells, the major increase was observed in the effector memory (EM) and in the CD45RA+ terminal effector T cell population (TEMRA CD8+) (Fig. 2D), whereas the other T cell populations, including naïve and central memory (CM), were nearly unaffected. These data are wholly aligned with recently published data13 showing that expansion of CD8+ cytotoxic T cells, including long lived, antigen-experienced T cells (CD45RA+), contribute to SARS-CoV-2 recovery in cancer patients. Intriguingly, in three patients with relapsed multiple myeloma enrolled in our single agent leflunomide phase 1 trial9, we observed that leflunomide elicited the same immune stimulatory effect on CD8+ cytotoxic T cells (unpublished data), further supporting an unexpected immune stimulatory role of this agent in cancer patients.

JISS-22-1241-fig6

Supplementary Figure 3: A) Line graph showing percentages of monocyte cells in PBMCs in both patients; B-C) Bar graphs showing percentages of monocyte cell immune sub-compartments, based on CD14 and CD16 expression; D-E) t-SNE heatmaps showing overall CD14 expression through the course of treatment in patients 1 and 2; F-G) Dot plots colored by CD14 channel for the signal of Classical, Intermediate and Non-Classical monocyte expression through the course of treatment. H-I) Bar graphs showing percentages of NKT cells compared to the total count PBMCs in Patients 1 (H) and 2 (I) over time, as measured by CyTOF; J-K) Bar graphs showing percentages of TCR-γδ cells compared to the total count PBMC in Patients 1 (J) and 2 (K) over time, as measured by CyTOF.

JISS-22-1241-fig7

Supplementary Figure 4: A-B-C) Line graph showing percentages of NK cells in both patients (A), and bar graphs showing percentages of early and mature NK cell immune sub-compartments in both patients at the indicated timepoints (B-C); D-E) Dot plots colored by CD56 channel for the signal of early and mature NK cell expression through the course of treatment.

Supplementary Table 3: CyTOF counts, Patient 1

Timepoint

Population

FCS Filename

Event count

Baseline

Leukocytes

20291_PB_Basline

151061

Baseline

Granulocytes

20291_PB_Basline

0

Baseline

PBMCs

20291_PB_Basline

151061

Baseline

B-cells

20291_PB_Basline

6986

Baseline

Naive B-cells

20291_PB_Basline

4515

Baseline

Total Memory B-cells

20291_PB_Basline

2471

Baseline

Memory B-cells

20291_PB_Basline

246

Baseline

Plasmablast

20291_PB_Basline

2160

Baseline

CD3- CD19-

20291_PB_Basline

85407

Baseline

CD56+ CD14+

20291_PB_Basline

18060

Baseline

NK-cells

20291_PB_Basline

7512

Baseline

Early NK

20291_PB_Basline

4185

Baseline

Mature NK

20291_PB_Basline

3322

Baseline

Monocytes ?

20291_PB_Basline

59803

Baseline

Classical Monocytes

20291_PB_Basline

45571

Baseline

Intermediate Monocytes

20291_PB_Basline

412

Baseline

Non-Classical Monocytes

20291_PB_Basline

1858

Baseline

CD14- CD16-

20291_PB_Basline

11712

Baseline

Myeloid Dendritic cells

20291_PB_Basline

1604

Baseline

Plasmacytoid Dendritic cells

20291_PB_Basline

47

Baseline

CD3+

20291_PB_Basline

58560

Baseline

NKT

20291_PB_Basline

1486

Baseline

T-cells

20291_PB_Basline

57072

Baseline

TCR g/d

20291_PB_Basline

620

Baseline

TCR a/b

20291_PB_Basline

56452

Baseline

CD8 T-cells

20291_PB_Basline

19802

Baseline

Effector CD8 T-cells

20291_PB_Basline

1220

Baseline

Naive CD8

20291_PB_Basline

7996

Baseline

CD8+ CD27-

20291_PB_Basline

5143

Baseline

Late differentiated CD8 T-cells

20291_PB_Basline

1650

Baseline

Temra CD8

20291_PB_Basline

3488

Baseline

CD8+ CD45RA- CD27+

20291_PB_Basline

5450

Baseline

CM CD8

20291_PB_Basline

2317

Baseline

CD8+ CD45RA- CD27+ CCR7-

20291_PB_Basline

3123

Baseline

EM CD8

20291_PB_Basline

1253

Baseline

Transitional memory CD8 T-cells

20291_PB_Basline

1870

Baseline

CD4 T-cells

20291_PB_Basline

35741

Baseline

CD4+ CD25+

20291_PB_Basline

1649

Baseline

Activated Effector CD4 T-cells

20291_PB_Basline

319

Baseline

Total Tregs

20291_PB_Basline

1330

Baseline

Memory Tregs

20291_PB_Basline

1301

Baseline

Naive Tregs

20291_PB_Basline

29

Baseline

CD4+ CD25-

20291_PB_Basline

34092

Baseline

CM CD4 T-cells

20291_PB_Basline

16059

Baseline

Naive CD4 T-cells

20291_PB_Basline

10541

Baseline

CD4+ CCR7- CD45RA?

20291_PB_Basline

7473

Baseline

EM CD4

20291_PB_Basline

5353

Baseline

Temra CD4

20291_PB_Basline

2120

D5

Leukocytes

20291_PB_D5

151061

D5

Granulocytes

20291_PB_D5

0

D5

PBMCs

20291_PB_D5

151061

D5

B-cells

20291_PB_D5

2667

D5

Naive B-cells

20291_PB_D5

1766

D5

Total Memory B-cells

20291_PB_D5

901

D5

Memory B-cells

20291_PB_D5

250

D5

Plasmablast

20291_PB_D5

605

D5

CD3- CD19-

20291_PB_D5

80611

D5

CD56+ CD14+

20291_PB_D5

12229

D5

NK-cells

20291_PB_D5

24543

D5

Early NK

20291_PB_D5

8525

D5

Mature NK

20291_PB_D5

15992

D5

Monocytes ?

20291_PB_D5

43783

D5

Classical Monocytes

20291_PB_D5

32035

D5

Intermediate Monocytes

20291_PB_D5

4817

D5

Non-Classical Monocytes

20291_PB_D5

4244

D5

CD14- CD16-

20291_PB_D5

2628

D5

Myeloid Dendritic cells

20291_PB_D5

547

D5

Plasmacytoid Dendritic cells

20291_PB_D5

31

D5

CD3+

20291_PB_D5

67731

D5

NKT

20291_PB_D5

5856

D5

T-cells

20291_PB_D5

61874

D5

TCR g/d

20291_PB_D5

1629

D5

TCR a/b

20291_PB_D5

60245

D5

CD8 T-cells

20291_PB_D5

36652

D5

Effector CD8 T-cells

20291_PB_D5

4553

D5

Naive CD8

20291_PB_D5

5435

D5

CD8+ CD27-

20291_PB_D5

21065

D5

Late differentiated CD8 T-cells

20291_PB_D5

5815

D5

Temra CD8

20291_PB_D5

15243

D5

CD8+ CD45RA- CD27+

20291_PB_D5

5613

D5

CM CD8

20291_PB_D5

1273

D5

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D5

4334

D5

EM CD8

20291_PB_D5

2579

D5

Transitional memory CD8 T-cells

20291_PB_D5

1755

D5

CD4 T-cells

20291_PB_D5

22510

D5

CD4+ CD25+

20291_PB_D5

856

D5

Activated Effector CD4 T-cells

20291_PB_D5

228

D5

Total Tregs

20291_PB_D5

628

D5

Memory Tregs

20291_PB_D5

617

D5

Naive Tregs

20291_PB_D5

11

D5

CD4+ CD25-

20291_PB_D5

21654

D5

CM CD4 T-cells

20291_PB_D5

8378

D5

Naive CD4 T-cells

20291_PB_D5

5390

D5

CD4+ CCR7- CD45RA?

20291_PB_D5

7874

D5

EM CD4

20291_PB_D5

4788

D5

Temra CD4

20291_PB_D5

3086

D8

Leukocytes

20291_PB_D8

151061

D8

Granulocytes

20291_PB_D8

0

D8

PBMCs

20291_PB_D8

151061

D8

B-cells

20291_PB_D8

2670

D8

Naive B-cells

20291_PB_D8

2093

D8

Total Memory B-cells

20291_PB_D8

577

D8

Memory B-cells

20291_PB_D8

384

D8

Plasmablast

20291_PB_D8

154

D8

CD3- CD19-

20291_PB_D8

76613

D8

CD56+ CD14+

20291_PB_D8

14437

D8

NK-cells

20291_PB_D8

12448

D8

Early NK

20291_PB_D8

4902

D8

Mature NK

20291_PB_D8

7536

D8

Monocytes ?

20291_PB_D8

49677

D8

Classical Monocytes

20291_PB_D8

40484

D8

Intermediate Monocytes

20291_PB_D8

1687

D8

Non-Classical Monocytes

20291_PB_D8

2666

D8

CD14- CD16-

20291_PB_D8

4737

D8

Myeloid Dendritic cells

20291_PB_D8

576

D8

Plasmacytoid Dendritic cells

20291_PB_D8

75

D8

CD3+

20291_PB_D8

71737

D8

NKT

20291_PB_D8

4110

D8

T-cells

20291_PB_D8

67625

D8

TCR g/d

20291_PB_D8

1291

D8

TCR a/b

20291_PB_D8

66334

D8

CD8 T-cells

20291_PB_D8

30916

D8

Effector CD8 T-cells

20291_PB_D8

3711

D8

Naive CD8

20291_PB_D8

6102

D8

CD8+ CD27-

20291_PB_D8

12227

D8

Late differentiated CD8 T-cells

20291_PB_D8

3473

D8

Temra CD8

20291_PB_D8

8754

D8

CD8+ CD45RA- CD27+

20291_PB_D8

8882

D8

CM CD8

20291_PB_D8

3057

D8

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D8

5814

D8

EM CD8

20291_PB_D8

2298

D8

Transitional memory CD8 T-cells

20291_PB_D8

3516

D8

CD4 T-cells

20291_PB_D8

34085

D8

CD4+ CD25+

20291_PB_D8

1257

D8

Activated Effector CD4 T-cells

20291_PB_D8

285

D8

Total Tregs

20291_PB_D8

972

D8

Memory Tregs

20291_PB_D8

958

D8

Naive Tregs

20291_PB_D8

14

D8

CD4+ CD25-

20291_PB_D8

32828

D8

CM CD4 T-cells

20291_PB_D8

15459

D8

Naive CD4 T-cells

20291_PB_D8

5126

D8

CD4+ CCR7- CD45RA?

20291_PB_D8

12220

D8

EM CD4

20291_PB_D8

8907

D8

Temra CD4

20291_PB_D8

3313

D14

Leukocytes

20291_PB_D14

151061

D14

Granulocytes

20291_PB_D14

0

D14

PBMCs

20291_PB_D14

151061

D14

B-cells

20291_PB_D14

897

D14

Naive B-cells

20291_PB_D14

765

D14

Total Memory B-cells

20291_PB_D14

132

D14

Memory B-cells

20291_PB_D14

104

D14

Plasmablast

20291_PB_D14

26

D14

CD3- CD19-

20291_PB_D14

58555

D14

CD56+ CD14+

20291_PB_D14

1185

D14

NK-cells

20291_PB_D14

15179

D14

Early NK

20291_PB_D14

4511

D14

Mature NK

20291_PB_D14

10650

D14

Monocytes ?

20291_PB_D14

42158

D14

Classical Monocytes

20291_PB_D14

21098

D14

Intermediate Monocytes

20291_PB_D14

10818

D14

Non-Classical Monocytes

20291_PB_D14

7742

D14

CD14- CD16-

20291_PB_D14

2534

D14

Myeloid Dendritic cells

20291_PB_D14

1199

D14

Plasmacytoid Dendritic cells

20291_PB_D14

800

D14

CD3+

20291_PB_D14

91559

D14

NKT

20291_PB_D14

5436

D14

T-cells

20291_PB_D14

86108

D14

TCR g/d

20291_PB_D14

1418

D14

TCR a/b

20291_PB_D14

84690

D14

CD8 T-cells

20291_PB_D14

46721

D14

Effector CD8 T-cells

20291_PB_D14

6100

D14

Naive CD8

20291_PB_D14

8287

D14

CD8+ CD27-

20291_PB_D14

17649

D14

Late differentiated CD8 T-cells

20291_PB_D14

4910

D14

Temra CD8

20291_PB_D14

12739

D14

CD8+ CD45RA- CD27+

20291_PB_D14

14706

D14

CM CD8

20291_PB_D14

3339

D14

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D14

11354

D14

EM CD8

20291_PB_D14

5743

D14

Transitional memory CD8 T-cells

20291_PB_D14

5611

D14

CD4 T-cells

20291_PB_D14

36245

D14

CD4+ CD25+

20291_PB_D14

1234

D14

Activated Effector CD4 T-cells

20291_PB_D14

165

D14

Total Tregs

20291_PB_D14

1069

D14

Memory Tregs

20291_PB_D14

939

D14

Naive Tregs

20291_PB_D14

130

D14

CD4+ CD25-

20291_PB_D14

35011

D14

CM CD4 T-cells

20291_PB_D14

16288

D14

Naive CD4 T-cells

20291_PB_D14

10797

D14

CD4+ CCR7- CD45RA?

20291_PB_D14

7899

D14

EM CD4

20291_PB_D14

4888

D14

Temra CD4

20291_PB_D14

3011

D21

Leukocytes

20291_PB_D21

151061

D21

Granulocytes

20291_PB_D21

0

D21

PBMCs

20291_PB_D21

151061

D21

B-cells

20291_PB_D21

607

D21

Naive B-cells

20291_PB_D21

564

D21

Total Memory B-cells

20291_PB_D21

43

D21

Memory B-cells

20291_PB_D21

37

D21

Plasmablast

20291_PB_D21

4

D21

CD3- CD19-

20291_PB_D21

68944

D21

CD56+ CD14+

20291_PB_D21

1396

D21

NK-cells

20291_PB_D21

26605

D21

Early NK

20291_PB_D21

7638

D21

Mature NK

20291_PB_D21

18946

D21

Monocytes ?

20291_PB_D21

40864

D21

Classical Monocytes

20291_PB_D21

17955

D21

Intermediate Monocytes

20291_PB_D21

6880

D21

Non-Classical Monocytes

20291_PB_D21

10871

D21

CD14- CD16-

20291_PB_D21

5147

D21

Myeloid Dendritic cells

20291_PB_D21

1701

D21

Plasmacytoid Dendritic cells

20291_PB_D21

1644

D21

CD3+

20291_PB_D21

81442

D21

NKT

20291_PB_D21

5272

D21

T-cells

20291_PB_D21

76163

D21

TCR g/d

20291_PB_D21

2945

D21

TCR a/b

20291_PB_D21

73218

D21

CD8 T-cells

20291_PB_D21

45223

D21

Effector CD8 T-cells

20291_PB_D21

10028

D21

Naive CD8

20291_PB_D21

5742

D21

CD8+ CD27-

20291_PB_D21

18462

D21

Late differentiated CD8 T-cells

20291_PB_D21

5045

D21

Temra CD8

20291_PB_D21

13414

D21

CD8+ CD45RA- CD27+

20291_PB_D21

11026

D21

CM CD8

20291_PB_D21

1706

D21

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D21

9310

D21

EM CD8

20291_PB_D21

6616

D21

Transitional memory CD8 T-cells

20291_PB_D21

2694

D21

CD4 T-cells

20291_PB_D21

26209

D21

CD4+ CD25+

20291_PB_D21

1477

D21

Activated Effector CD4 T-cells

20291_PB_D21

206

D21

Total Tregs

20291_PB_D21

1271

D21

Memory Tregs

20291_PB_D21

1168

D21

Naive Tregs

20291_PB_D21

103

D21

CD4+ CD25-

20291_PB_D21

24732

D21

CM CD4 T-cells

20291_PB_D21

8595

D21

Naive CD4 T-cells

20291_PB_D21

6552

D21

CD4+ CCR7- CD45RA?

20291_PB_D21

9574

D21

EM CD4

20291_PB_D21

3545

D21

Temra CD4

20291_PB_D21

6029

D28

Leukocytes

20291_PB_D28

151061

D28

Granulocytes

20291_PB_D28

0

D28

PBMCs

20291_PB_D28

151061

D28

B-cells

20291_PB_D28

476

D28

Naive B-cells

20291_PB_D28

448

D28

Total Memory B-cells

20291_PB_D28

28

D28

Memory B-cells

20291_PB_D28

21

D28

Plasmablast

20291_PB_D28

7

D28

CD3- CD19-

20291_PB_D28

77699

D28

CD56+ CD14+

20291_PB_D28

1618

D28

NK-cells

20291_PB_D28

25055

D28

Early NK

20291_PB_D28

9258

D28

Mature NK

20291_PB_D28

15773

D28

Monocytes ?

20291_PB_D28

50952

D28

Classical Monocytes

20291_PB_D28

22859

D28

Intermediate Monocytes

20291_PB_D28

10833

D28

Non-Classical Monocytes

20291_PB_D28

12452

D28

CD14- CD16-

20291_PB_D28

4828

D28

Myeloid Dendritic cells

20291_PB_D28

1566

D28

Plasmacytoid Dendritic cells

20291_PB_D28

1749

D28

CD3+

20291_PB_D28

72834

D28

NKT

20291_PB_D28

3806

D28

T-cells

20291_PB_D28

69023

D28

TCR g/d

20291_PB_D28

4092

D28

TCR a/b

20291_PB_D28

64931

D28

CD8 T-cells

20291_PB_D28

39848

D28

Effector CD8 T-cells

20291_PB_D28

12908

D28

Naive CD8

20291_PB_D28

4526

D28

CD8+ CD27-

20291_PB_D28

14265

D28

Late differentiated CD8 T-cells

20291_PB_D28

4261

D28

Temra CD8

20291_PB_D28

10004

D28

CD8+ CD45RA- CD27+

20291_PB_D28

8160

D28

CM CD8

20291_PB_D28

1624

D28

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D28

6524

D28

EM CD8

20291_PB_D28

4572

D28

Transitional memory CD8 T-cells

20291_PB_D28

1952

D28

CD4 T-cells

20291_PB_D28

23246

D28

CD4+ CD25+

20291_PB_D28

1010

D28

Activated Effector CD4 T-cells

20291_PB_D28

128

D28

Total Tregs

20291_PB_D28

882

D28

Memory Tregs

20291_PB_D28

835

D28

Naive Tregs

20291_PB_D28

47

D28

CD4+ CD25-

20291_PB_D28

22236

D28

CM CD4 T-cells

20291_PB_D28

8490

D28

Naive CD4 T-cells

20291_PB_D28

5365

D28

CD4+ CCR7- CD45RA?

20291_PB_D28

8371

D28

EM CD4

20291_PB_D28

3362

D28

Temra CD4

20291_PB_D28

5009

D40

Leukocytes

20291_PB_D40

151061

D40

Granulocytes

20291_PB_D40

0

D40

PBMCs

20291_PB_D40

151061

D40

B-cells

20291_PB_D40

689

D40

Naive B-cells

20291_PB_D40

425

D40

Total Memory B-cells

20291_PB_D40

264

D40

Memory B-cells

20291_PB_D40

38

D40

Plasmablast

20291_PB_D40

206

D40

CD3- CD19-

20291_PB_D40

69036

D40

CD56+ CD14+

20291_PB_D40

1481

D40

NK-cells

20291_PB_D40

26394

D40

Early NK

20291_PB_D40

8310

D40

Mature NK

20291_PB_D40

18050

D40

Monocytes ?

20291_PB_D40

41087

D40

Classical Monocytes

20291_PB_D40

17194

D40

Intermediate Monocytes

20291_PB_D40

8146

D40

Non-Classical Monocytes

20291_PB_D40

11709

D40

CD14- CD16-

20291_PB_D40

4072

D40

Myeloid Dendritic cells

20291_PB_D40

1331

D40

Plasmacytoid Dendritic cells

20291_PB_D40

644

D40

CD3+

20291_PB_D40

81280

D40

NKT

20291_PB_D40

2476

D40

T-cells

20291_PB_D40

78802

D40

TCR g/d

20291_PB_D40

3016

D40

TCR a/b

20291_PB_D40

75786

D40

CD8 T-cells

20291_PB_D40

40159

D40

Effector CD8 T-cells

20291_PB_D40

10555

D40

Naive CD8

20291_PB_D40

6122

D40

CD8+ CD27-

20291_PB_D40

14136

D40

Late differentiated CD8 T-cells

20291_PB_D40

3103

D40

Temra CD8

20291_PB_D40

11015

D40

CD8+ CD45RA- CD27+

20291_PB_D40

9392

D40

CM CD8

20291_PB_D40

2300

D40

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D40

7082

D40

EM CD8

20291_PB_D40

4816

D40

Transitional memory CD8 T-cells

20291_PB_D40

2266

D40

CD4 T-cells

20291_PB_D40

33775

D40

CD4+ CD25+

20291_PB_D40

952

D40

Activated Effector CD4 T-cells

20291_PB_D40

134

D40

Total Tregs

20291_PB_D40

818

D40

Memory Tregs

20291_PB_D40

749

D40

Naive Tregs

20291_PB_D40

69

D40

CD4+ CD25-

20291_PB_D40

32823

D40

CM CD4 T-cells

20291_PB_D40

12546

D40

Naive CD4 T-cells

20291_PB_D40

7278

D40

CD4+ CCR7- CD45RA?

20291_PB_D40

12963

D40

EM CD4

20291_PB_D40

7191

D40

Temra CD4

20291_PB_D40

5772

D56

Leukocytes

20291_PB_D56

151061

D56

Granulocytes

20291_PB_D56

0

D56

PBMCs

20291_PB_D56

151061

D56

B-cells

20291_PB_D56

1205

D56

Naive B-cells

20291_PB_D56

805

D56

Total Memory B-cells

20291_PB_D56

400

D56

Memory B-cells

20291_PB_D56

66

D56

Plasmablast

20291_PB_D56

292

D56

CD3- CD19-

20291_PB_D56

85540

D56

CD56+ CD14+

20291_PB_D56

1927

D56

NK-cells

20291_PB_D56

30493

D56

Early NK

20291_PB_D56

9146

D56

Mature NK

20291_PB_D56

21311

D56

Monocytes ?

20291_PB_D56

53028

D56

Classical Monocytes

20291_PB_D56

26053

D56

Intermediate Monocytes

20291_PB_D56

8935

D56

Non-Classical Monocytes

20291_PB_D56

14216

D56

CD14- CD16-

20291_PB_D56

3861

D56

Myeloid Dendritic cells

20291_PB_D56

1022

D56

Plasmacytoid Dendritic cells

20291_PB_D56

1979

D56

CD3+

20291_PB_D56

64265

D56

NKT

20291_PB_D56

2541

D56

T-cells

20291_PB_D56

61720

D56

TCR g/d

20291_PB_D56

3596

D56

TCR a/b

20291_PB_D56

58124

D56

CD8 T-cells

20291_PB_D56

30821

D56

Effector CD8 T-cells

20291_PB_D56

7494

D56

Naive CD8

20291_PB_D56

7316

D56

CD8+ CD27-

20291_PB_D56

10681

D56

Late differentiated CD8 T-cells

20291_PB_D56

2670

D56

Temra CD8

20291_PB_D56

8011

D56

CD8+ CD45RA- CD27+

20291_PB_D56

5350

D56

CM CD8

20291_PB_D56

1911

D56

CD8+ CD45RA- CD27+ CCR7-

20291_PB_D56

3430

D56

EM CD8

20291_PB_D56

2230

D56

Transitional memory CD8 T-cells

20291_PB_D56

1200

D56

CD4 T-cells

20291_PB_D56

25412

D56

CD4+ CD25+

20291_PB_D56

1293

D56

Activated Effector CD4 T-cells

20291_PB_D56

114

D56

Total Tregs

20291_PB_D56

1179

D56

Memory Tregs

20291_PB_D56

1084

D56

Naive Tregs

20291_PB_D56

95

D56

CD4+ CD25-

20291_PB_D56

24119

D56

CM CD4 T-cells

20291_PB_D56

8984

D56

Naive CD4 T-cells

20291_PB_D56

8027

D56

CD4+ CCR7- CD45RA?

20291_PB_D56

7095

D56

EM CD4

20291_PB_D56

3532

D56

Temra CD4

20291_PB_D56

3563

Supplementary Table 4: CyTOF Counts, Patient 2

Population

Timepoint

FCS Filename

Event count

PBMCs

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

95678

PBMCs

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

95678

PBMCs

D8

Pt2_D8_PBMC_merge1

95678

PBMCs

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

95678

PBMCs

D21

Pt.#2_D21_PBMC_IRB20291_MDIPA_10-13-2021_1

95678

PBMCs

D28

Pt.#2_D28_PBMC_IRB20291_MDIPA_10-13-2021_1

95678

B-cells

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

12212

B-cells

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

5966

B-cells

D8

Pt2_D8_PBMC_merge1

3561

B-cells

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

4293

B-cells

D21

Pt.#2_D21_PBMC_IRB20291_MDIPA_10-13-2021_1

5977

B-cells

D28

Pt.#2_D28_PBMC_IRB20291_MDIPA_10-13-2021_1

2283

Naive B-cells

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

12020

Naive B-cells

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

5661

Naive B-cells

D8

Pt2_D8_PBMC_merge1

3528

Naive B-cells

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

3865

Naive B-cells

D21

Pt.#2_D21_PBMC_IRB20291_MDIPA_10-13-2021_1

5140

Naive B-cells

D28

Pt.#2_D28_PBMC_IRB20291_MDIPA_10-13-2021_1

2232

Total Memory B-cells

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

191

Total Memory B-cells

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

305

Total Memory B-cells

D8

Pt2_D8_PBMC_merge1

32

Total Memory B-cells

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

427

Total Memory B-cells

D21

Pt.#2_D21_PBMC_IRB20291_MDIPA_10-13-2021_1

835

Total Memory B-cells

D28

Pt.#2_D28_PBMC_IRB20291_MDIPA_10-13-2021_1

51

Plasmablast

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

47

Plasmablast

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

5

Plasmablast

D8

Pt2_D8_PBMC_merge1

4

Plasmablast

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

8

Plasmablast

D21

Pt.#2_D21_PBMC_IRB20291_MDIPA_10-13-2021_1

23

Plasmablast

D28

Pt.#2_D28_PBMC_IRB20291_MDIPA_10-13-2021_1

7

NK-cells

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

9448

NK-cells

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

6875

NK-cells

D8

Pt2_D8_PBMC_merge1

11434

NK-cells

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

4670

NK-cells

D21

Pt.#2_D21_PBMC_IRB20291_MDIPA_10-13-2021_1

7353

NK-cells

D28

Pt.#2_D28_PBMC_IRB20291_MDIPA_10-13-2021_1

5770

Early NK

Screening

Pt#2_Screening_PBMC_IRB20291_MDPA_11-10-2021_1_concat

3623

Early NK

D4

Pt.#2_D4_PBMC_IRB20291_MDIPA_10-13-2021_1

3130

Early NK

D8

Pt2_D8_PBMC_merge1

4462

Early NK

D14

Pt.#2_D14_PBMC_IRB20291_MDIPA_10-13-2021_1

1841

Although changes in the percentage of total CD4+ T cells differed among the two patients, both patients showed an increase in CD4+ TEMRA cells, while instead an increase in CM and EM CD4+ T cells was only present in the second patient (Fig. 2D and Sup. Fig. 2H-J).

An increase in pro-inflammatory CD16+ cells, such as intermediate and non-classical monocytes, was additionally observed in both patients, but the classical monocyte population remained almost unchanged (Sup. Fig. 3A-G). In line with our observation that leflunomide appears to generally increase anti-viral T cell responses, we also noted a consistent increase among the two patients in total NKT (Sup. Fig. 3H-I) and TCR yδ cells (Sup. Fig. 3J-K), which play a pivotal role in countering viral infection14. A robust increase in the NK cell population, specifically in mature NK cells, over the course of the study was noted in the first patient (Sup. Fig. 4A-E).

Consistent with the decrease in the B cell population, the immunoglobulin profile showed a decrease in total IgM levels in the first patient (Sup. Fig. 5A), an effect that was previously associated with a relatively rapid recovery from COVID-1915. This effect was observed seven days after the patient began leflunomide treatment, while total IgG levels remained mostly unchanged (Sup. Fig. 5B). A decrease in IgM levels was instead not observed in the second patient (Sup. Fig. 5C-D).

JISS-22-1241-fig8

Supplementary Figure 5: A-B-C-D) Longitudinal evaluation of serum IgM (A,C) and IgG (B,D) immunoglobulin reactive to SARS-CoV-2 nucleoprotein (N), receptor-binding domain (RBD), and spike (S) collected from the patients at baseline (time zero), through the course of leflunomide treatment (up to 14 days) and follow up (up to 56 days); E-F) Longitudinal evaluation of serum cytokines IL-2R through the treatment course (14 days) and follow up appointments (up to day 56).

A cytokine array showed that leflunomide treatment decreased soluble IL2R (Sup. Fig. 5E-F); high levels are regularly associated with inhibition of CD8+ cytotoxic T cells and high rates of hospitalization and mortality16.

Consistent with previously published data, here we show that leflunomide affects B cell proliferation17, and for the first time report that leflunomide stimulates anti-viral activity by promoting innate immunity. Our patients treated on a leflunomide protocol had rapid improvements that were coupled with temporal and favorable changes in immunologic response. An important limitation is the small number of patients enrolled to date; however, the data align with two pilot studies comparing leflunomide to leflunomide plus institutional standard of care, one showing that leflunomide plus standard of care conferred favorable SARS-CoV-2 clearance and hospital discharge10, and the other demonstrating a shorter duration of viral shedding and a reduction in C-reactive protein18. A third study assigned patients to either leflunomide or leflunomide plus IFNα and found no statistically significant difference in terms of duration of viral shedding or length of hospital stay19. However, the dosing used was lower than in our study, and it is possible that the enhancement of IFNα signaling by leflunomide precludes additional benefit of IFNα intervention. Finally, two separate case series described patients taking teriflunomide for multiple sclerosis who contracted COVID-19 but experienced self-limiting infection20,21.

Conclusion

We used immunologic correlative experiments to describe the immune response of two patients with severe COVID-19 who were treated with the immune modulator leflunomide. Further study is warranted to more definitely address the effectiveness of leflunomide in the treatment of COVID-19, especially in patients with cancer. Leflunomide treatment might also be relevant for patients who are immunocompromised due to factors other than cancer, but additional evidence is needed. With ongoing COVID-19 transmission, which can overcome precautions such as hand washing and social distancing,22 and occurrence of breakthrough infections in vaccinated individuals, therapeutic agents that target both the virus and host inflammatory response would be helpful despite the availability of currently approved anti-viral agents. Furthermore, from an access to care perspective, especially in resource-limited areas, an inexpensive, readily available, effective drug with existing safety data in humans is relevant in the real-world setting.

Acknowledgments

We are grateful to the Pathogen & Microbiome Division at TGen North for the sequencing of SARS-CoV-2 strains. The Biostatistics Core at City of Hope was supported by the National Institutes of Health under award number P30CA033572. This research was also in part supported by a P30CA033572 Supplement. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. We also acknowledge support from the Norman and Sadie Lee Foundation.

Conflict of Interest

The authors declare that they have no competing interests that are relevant to this study.

References

  1. Priyanka, Choudhary OP, Singh I. Protective immunity against COVID-19: Unravelling the evidences for humoral vs. cellular components. Travel Med Infect Dis. 2021; 39: 101911.
  2. Davis JP, Cain GA, Pitts WJ, et al. The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase. Biochemistry. 1996; 35(4): 1270-1273.
  3. Baumann P, Mandl-Weber S, Völkl A, et al. Dihydroorotate dehydrogenase inhibitor A771726 (leflunomide) induces apoptosis and diminishes proliferation of multiple myeloma cells. Mol Cancer Ther. 2009; 8(2): 366-375.
  4. Romano M, Ruggiero A, Squeglia F, et al. A Structural View of SARS-CoV-2 RNA Replication Machinery: RNA Synthesis, Proofreading and Final Capping. Cells. 2020; 9(5).
  5. Parekh JM, Vaghela RN, Sutariya DK, et al. Chromatographic separation and sensitive determination of teriflunomide, an active metabolite of leflunomide in human plasma by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2010; 878(24): 2217-2225.
  6. Xiong R, Zhang L, Li S, et al. Novel and potent inhibitors targeting DHODH, a rate-limiting enzyme in de novo pyrimidine biosynthesis, are broad-spectrum antiviral against RNA viruses including newly emerged coronavirus SARS-CoV-2. bioRxiv. 2020:2020.2003.2011.983056.
  7. Wang E, Jan AS, Doan VP, et al. Leflunomide therapy for refractory cytomegalovirus infections in hematopoietic stem cell transplant recipients. J Oncol Pharm Pract. 2019; 25(7): 1731-1737.
  8. Williams JW, Javaid B, Kadambi PV, et al. Leflunomide for polyomavirus type BK nephropathy. N Engl J Med. 2005; 352(11): 1157-1158.
  9. Rosenzweig M, Palmer J, Tsai NC, et al. Repurposing leflunomide for relapsed/refractory multiple myeloma: a phase 1 study. Leuk Lymphoma. 2020; 61(7): 1669-1677.
  10. Wang Q, Guo H, Li Y, et al. Efficacy and Safety of Leflunomide for Refractory COVID-19: A Pilot Study. Front Pharmacol. 2021; 12: 581833.
  11. Siemasko KF, Chong AS, Williams JW, et al. Regulation of B cell function by the immunosuppressive agent leflunomide. Transplantation. 1996; 61(4): 635-642.
  12. Breedveld FC, Dayer JM. Leflunomide: mode of action in the treatment of rheumatoid arthritis. Ann Rheum Dis. 2000; 59(11): 841-849.
  13. Bange EM, Han NA, Wileyto P, et al. CD8(+) T cells contribute to survival in patients with COVID-19 and hematologic cancer. Nat Med. 2021; 27(7): 1280-1289.
  14. Sabbaghi A, Miri SM, Keshavarz M, et al. Role of γδ T cells in controlling viral infections with a focus on influenza virus: implications for designing novel therapeutic approaches. Virol J. 2020; 17(1): 174.
  15. Legros V, Denolly S, Vogrig M, et al. A longitudinal study of SARS-CoV-2-infected patients reveals a high correlation between neutralizing antibodies and COVID-19 severity. Cell Mol Immunol. 2021; 18(2): 318-327.
  16. Jang HJ, Leem AY, Chung KS, et al. Soluble IL-2R Levels Predict in-Hospital Mortality in COVID-19 Patients with Respiratory Failure. J Clin Med. 2021; 10(18).
  17. van der Heijden EH, Hartgring SA, Kruize AA, et al. Additive immunosuppressive effect of leflunomide and hydroxychloroquine supports rationale for combination therapy for Sjögren's syndrome. Expert Rev Clin Immunol. 2019; 15(7): 801-808.
  18. Hu K, Wang M, Zhao Y, et al. A Small-Scale Medication of Leflunomide as a Treatment of COVID-19 in an Open-Label Blank-Controlled Clinical Trial. Virologica Sinica. 2020; 35(6): 725-733.
  19. Wang M, Zhao Y, Hu W, et al. Treatment of Coronavirus Disease 2019 Patients With Prolonged Postsymptomatic Viral Shedding With Leflunomide: A Single-center Randomized Controlled Clinical Trial. Clinical Infectious Diseases. 2020; 73(11): e4012-e4019.
  20. Maghzi AH, Houtchens MK, Preziosa P, et al. COVID-19 in teriflunomide-treated patients with multiple sclerosis. Journal of Neurology. 2020; 267(10): 2790-2796.
  21. Mantero V, Baroncini D, Balgera R, et al. Mild COVID‑19 infection in a group of teriflunomide‑treated patients with multiple sclerosis. Journal of Neurology. 2021; 268(6): 2029-2030.
  22. Priyanka, Choudhary OP, Singh I, et al. Aerosol transmission of SARS-CoV-2: The unresolved paradox. Travel Med Infect Dis. 2020; 37: 101869.
 

Article Info

Article Notes

  • Published on: January 20, 2023

Keywords

  • Leflunomide
  • COVID-19
  • Breast cancer
  • Clinical trial
  • Drug repurposing

*Correspondence:

Steven Rosen,
Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA;
Email: srosen@coh,org
Sanjeet Dadwal,
Department of Medicine, Division of Infectious Disease, City of Hope, Duarte, CA, USA;
Email: sdadwal@coh.org
Flavia Pichiorri,
Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA;
Email: fpichiorri@coh.org

Copyright: ©2023 Rosen S. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License.

Copyright: ©2023 Dadwal S. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License.

Copyright: ©2023 Pichiorri F. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License.