Trained Immunity: RoadMap for drug discovery and development

  1. Jelmer H van Puffelen  Is a corresponding author
  2. Callum Campbell
  3. Irene Gander-Meisterernst
  4. Johanna Holldack
  5. Pauline T Lukey
  1. Kupando GmbH, Germany
  2. Target to Treatment Consulting Ltd, Stevenage Bioscience Catalyst, United Kingdom
10 figures and 5 tables

Figures

Illustration of Trained Immunity with a focus on the induction of Trained Immunity via Toll-like receptors, Dectin-1, and Nod-like receptor activation.

These receptors are triggered by pathogen-associated molecular patterns (PAMP), and innate immune cells such as monocytes and macrophages are activated to produce inflammatory cytokines. Dectin-1 is the receptor that recognizes β-glucan and thereby initiates β-glucan-induced Trained Immunity. NOD-like receptors recognize Bacillus Calmette-Guérin (BCG) and thereby mediate BCG-induced Trained Immunity. Toll-like receptors recognize various types of PAMPs and can also mediate Trained Immunity (a). The primary immune response to infection is characterized by cytokine production and antigen presentation (b). The primary innate immune response dissipates, and the innate immune cell returns to baseline activation state; however, epigenetic changes, driven by metabolic reprogramming, persist at the chromatin level. This epigenetic reprogramming underlies the induction of Trained Immunity (c). Subsequent restimulation with an unrelated pathogen or immunological trigger initiates a Trained Immunity response that is characterized by adaptive and enhanced effector functions such as increased cytokine production, enhanced antigen presentation, and increased phagocytosis (d). As a secondary effect of Trained Immunity, this may lead to enhanced adaptive immune responses.

Relevant domains for Trained Immunity-targeted drug discovery and development.

Five relevant domains for investigation are illustrated. Epigenetic, metabolic, and inflammatory changes, as well as differentiation and memory, are important for drug discovery and development. (Icons created by the Noun project: https://thenounproject.com/.)

Simplified overview of a drug discovery and development pipeline.

The main stages of drug discovery and development are included in the chevrons from target identification and validation through Phase 3 clinical trials. Each stage has a brief description in the boxes below the chevrons. The diamond shapes represent significant milestone achievements along the pathway. These include the selection of the single candidate that will be progressed to the clinic and the submission of the data package and clinical protocol to the regulators for approval to enter the clinic (FDA: open an IND application; EMA: CTA). Clinical proof of concept (PoC) is achieved when clinical effects of the new drug are demonstrated in patients (usually Phase 2a). The ‘commit to Phase 3’ meeting with regulators occurs when the therapeutic dose has been identified in Phase 2b and has been ratified by the regulators. At the end of Phase 3, the entirety of the data is submitted to the regulators for approval to make the new drug available to patients.

All steps from research to development up to the start of clinical development can be supported by AI.

In an iterative process, data is generated in the laboratory as shown in the upper half of the figure in gray, with AI offering valuable input at each stage as shown in the lower half of the figure in blue: From target identification to the selection of nonclinical in vitro and in vivo models, and finally, the identification of patient subsets and biomarker and outcome assessment once a drug candidate progresses to the clinical phase. Once the lead has been identified, development incorporates stringent adherence to development standards such as the GLP, GMP, and GCP guidelines and standards. Regulatory advice needs to be included to streamline drug development and entry into Phase 1 and to start building a target product profile. Drug discovery and development in Trained Immunity seeks to leverage and modify the innate immune system to achieve a durable and balanced immune response.

Illustration of potential types of models to assess Trained Immunity during the translational phase of drug development.
Types and applications of biomarkers (icons from the Noun project).
Discovery and development of biomarkers in parallel with the discovery and development of the drug.
Overview of a possible in vitro study design to investigate the activity of a test compound in a model of Trained Immunity.

A stimulus to prime innate immunity may be added to the model and endpoints measured (cytokines, metabolism, and epigenetics). Over time, the effect of the priming stimulus fades and most of the endpoints return to baseline levels (the exception being epigenetics, where the modifications may be longer-lived). The test compound may be added at various concentrations at the same time as the secondary challenge. Subsequent effects on cytokines, metabolism, and epigenetics may be measured. The basal response (measured in the absence of treatment) may be compared to the posttreatment response. The compound may decrease the trained immune response, or it may augment the response.

Selected clinical trials investigating the modification and regulation of Trained Immunity.

(a) Trials investigating induction of Trained Immunity for therapeutic benefit, (b) trials investigating modulation of Trained Immunity for therapeutic benefit, and (c) trials investigating inhibition of Trained Immunity for therapeutic benefit.

RoadMap to discovery and development of molecules that are designed to provide clinical benefit in indications relevant to Trained Immunity.

Tables

Table 1
Summary of the main classes of drug targets that target Trained Immunity and the main drug development domains they modulate.
Target classMain affected drug development domain
Pattern recognition receptors
Toll-like receptors (TLRs) (Alexopoulou and Irla, 2025)
NOD-like receptors (NLRs) (Kleinnijenhuis et al., 2012)
C-type lectin receptors (CLRs) (Moorlag et al., 2020; Moerings et al., 2021)
Epigenetic
Metabolic
Differentiation
Inflammatory
Memory
Cytokines and cytokine receptors
IL-1β and IL-1R (Moorlag et al., 2020; Teufel et al., 2022)
IL-4 and IL-4R (Schrijver et al., 2023)
Metabolic
Inflammation
Differentiation
Epigenetic enzymes
Histone acetyltransferases (Ziogas et al., 2025; Fanucchi et al., 2021),
Histone deacetylases (Cheng et al., 2014; Mourits et al., 2021a),
Histone methyltransferases (Keating et al., 2020b; Mourits et al., 2021b)
Histone demethylases (Arts et al., 2016)
Lactyltransferase (p300) (Ziogas et al., 2025)
Metabolic
Epigenetic
Memory
Metabolism
Hexokinase (Cheng et al., 2014)
Succinate dehydrogenase (Domínguez-Andrés et al., 2019)
Acetyl-CoA carboxylase (Arts et al., 2016)
Glutaminase (Scarpa et al., 2025)
Metabolic
Table 2
Trained Immunity-regulating compounds.
Description of Trained Immunity regulating compoundTypeTrained Immunity targetInducing or inhibiting Trained Immunity*Cellular location of Trained Immunity targetStatusReference
BCG vaccineLive-attenuated vaccineNOD2 receptor, TLR2, TLR4InducingIntracellularMarketedMoorlag et al., 2024
MV130Whole heat-inactivated bacteria (90% Gram-positive, 10% Gram-negative)Combination of TLRs and NLRsInducingIntracellular and extracellularIn developmentBrandi et al., 2022
MDP combined with HPV E7 peptide encapsulated by polylactic-co-glycolic acid PLGA nanoparticlesNanoparticleNOD2 receptorInducingIntracellularExperimentalLi et al., 2023
MTP10-HDLNanoparticleNOD2 receptorInducingIntracellularIn developmentPriem et al., 2020
PEG-PDLLA polymersome containing β-glucanPolymersomeDectin-1InducingExtracellular/transmembrane receptorIn developmentWauters et al., 2024
PLGA nanoparticles containing β-glucanNanoparticleDectin-1InducingExtracellular/transmembrane receptorExperimentalAjit et al., 2022
Saccharomyces cerevisiae-derived whole glucan particles containing β-glucanSonicated yeast particlesDectin-1InducingExtracellular/transmembrane receptorExperimentalHorneck Johnston et al., 2024
BIX‐01294Small moleculeG9a histone methyltransferaseInducingNucleoplasmExperimentalMourits et al., 2021b
Monophosphoryl lipid A (MPLA)Modified lipidTLR4InducingExtracellular/transmembrane receptorExperimentalOwen et al., 2022
Oxidized low-density lipoprotein (OxLDL)LipoproteinTLR4InducingExtracellular/transmembrane receptorExperimentalBekkering et al., 2014
CpG-ODNOligonucleotideTLR9InducingIntracellularExperimentalOwen et al., 2022
β-Glucan in arabinoxylanHemicelluloseComplement receptor 3 (CR3)InducingExtracellular/transmembraneExperimentalMoerings et al., 2022; Patin et al., 2019
Fusion protein of apolipoprotein A1 and IL4Lipid nanoparticleIL-4 receptor (type 1 and type 2)InducingExtracellular/transmembraneIn developmentSchrijver et al., 2023
AnakinraRecombinant proteinIL-1 receptorInhibitingExtracellularMarketedFlores-Gomez et al., 2024; Mitroulis et al., 2018; Ciarlo et al., 2020
CanakinumabMonoclonal antibodyIL-1β protein inhibitorInhibitingExtracellularMarketedXu et al., 2025
5′-Deoxy-5′methylthio adenosine (MTA)Synthetic organic compoundHistone methyltransferase inhibitorInhibitingNucleoplasmExperimentalQuintin et al., 2012
ResveratrolNatural polyphenolSirtuin 1 (histone deacetylase) activatorInducingNucleoplasmMarketed (as supplement)Mourits et al., 2021a; Bulut et al., 2025; Wang et al., 2021a
  1. *

    The aim of inducing or inhibiting Trained Immunity is to change the inflammatory state in a certain (disease) setting. Depending on the clinical indication, modulation of Trained Immunity responses can either boost or diminish inflammatory responses. For example, boosting immune responses via Trained Immunity can change the equilibrium during immunoparalysis. On the contrary, in a chronic hyperinflammatory state, inhibiting Trained Immunity responses could potentially serve as a tool to change the equilibrium the other way around.

Table 3
Examples of biomarkers and models of Trained Immunity.
Domains of Trained ImmunityKey markersAssayContextModelsExample references
EpigeneticsHistone methylation H3K4me3 (active promoters), Histone acetylation H3K27ac (enhancers), H3K9me3 (repressive epigenetic marks)
Chromatin accessibility
ChIP-seq, CUT&RUN, ATAC-seq, ChIP-PCRTrained Immunity induces chromatin remodeling at cytokine and metabolic gene lociIn vitro monocyte training (e.g. with β-glucan or BCG), in vitro training of mouse bone marrow-derived macrophagesYoung et al., 2011
Meers et al., 2019

Saeed et al., 2014
MetabolicGlycolysis: ↑ lactate, ↑ Glut1 expression
TCA cycle: Itaconate, succinate, fumarate accumulation
mTOR/HIF-1α activation
reactive oxygen species (ROS)
NAD+ metabolism
Seahorse (ECAR, OCR), metabolomics (LC-MS), western blot for pathway markers HK2, LDHA (glycolysis), HIF-1α, mTOR, AMPK, SDH (succinate dehydrogenase)
ROS assay
Pathways alteredIn vitro Trained Immunity models with monocytes or PBMCs, both with mouse and human cellsStefanoni, 2023
Wang et al., 2021b
Mourits et al., 2021a
Wu et al., 2024
InflammatoryIL-6, TNF-α, IL-1β, IL-10, IL-18, IFN-γELISA, Luminex, multiplex bead-based assaysCytokine Production after stimulation with unrelated secondary ligands (e.g. LPS, Pam3CSK4), trained cells show enhanced cytokine productionIn vitro monocyte and PBMC Trained Immunity assays, both with mouse and human cells. Primary human and mouse cells from in vivo studies can be used for ex vivo restimulation assaysMoorlag et al., 2020
Smith et al., 2017
DifferentiationMonocytes/macrophages: CD11b, CD14, CD16, CD80, CD86, HLA-DR
NK cells: CD69, NKG2D, CD107a (degranulation)
Flow cytometry, mass cytometry (CyTOF)Cell Surface and Activation Markers
Trained cells may show altered expression profiles reflecting activation or increased antigen presentation
Human PBMCs, mouse peritoneal, or splenic macrophagesGill et al., 2022
Zhang et al., 2021
MemoryPersistence: Epigenetic and transcriptional changes lasting weeks to months

Bone marrow signatures: Myeloid progenitor reprogramming
Clinical: BCG-vaccinated individuals show altered monocyte and cytokine profiles even after 3–12 months
See above examplesLongitudinal Biomarkers of Trained Immunity in in vivo studiesEx vivo, mouse models and human modelsBomans et al., 2018
Cassone, 2018

Kuznetsova et al., 2020
Table 4
Selected clinical trials investigating interventions in the context of Trained Immunity.
(a) Trials investigating induction of Trained Immunity for therapeutic benefit
1NCT IDNCT06257212
TitleLive Vaccines and Innate Immunity Training in COPD
Dates2024/02/28 to 2025/09
PhasePhase 4
Enrolment60 (Estimated)
Condition(s)COPD
Intervention(s)BCG vaccine
MMR vaccine
Primary OutcomeInnate immune training measured by fold-changes in cytokine production capacity of innate immune cells following pro-inflammatory stimulation. Measured from inclusion in the trial to 4 months’ post-inclusion. Cytokines include: IL-1β, IL-10, TNF-α, IFN-γ
2NCT IDNCT06266754
TitleThe Non-Specific Immunological Effects of Providing Oral Polio Vaccine to Seniors in Guinea-Bissau
Dates2024/01/29 to 2024/12/31
PhasePhase 4
Enrolment80 (Estimated)
Condition(s)Vaccine Reaction
Intervention(s)Oral Polio vaccine
Primary Outcome
  1. Levels of proinflammatory cytokines (including IL1-β, TNF-α, IFN-γ) after stimulation of PBMCs with non-OPV antigens and mitogens 1 month after intervention

  2. Levels of plasma markers of systemic inflammation (e.g. TWEAK and SIRT2) 1 month after intervention

  3. Investigating epigenetic changes in PBMCs by single-cell ATAC-sequencing and whole-genome methylation assays 1 month after intervention

  4. Investigate transcriptional effects on immune cells by single-cell RNA-sequencing 1 month after intervention. Identifying proportions of immune cell subsets

3NCT IDNCT05208060
TitleStudy to Evaluate the Ability of Sublingual MV130 to Induce the Expression of Trained Immunity in Peripheral Blood Cells
Dates2023/09/01 to 2025/12/31
PhasePhases 1 and 2
Enrolment48 (Estimated)
Condition(s)Immune Response
Intervention(s)MV130 vaccine
Primary OutcomeIncrease in ex vivo PBMCs cytokine response (TNF-α, IL-6, IL-1β) to secondary restimulation compared to placebo at days 15, 45, and 70 with respect to baseline
Selected Secondary Outcomes relevant to Trained Immunity
  1. Epigenetic and metabolic changes in purified monocytes from PBMCs, including specific Trained Immunity-associated miRNAs (miR155, miR146, miR21), lactate production, glucose consumption, and mitochondrial activity at day 45 with respect to baseline

  2. Change in proportions of immune cells (including T cells, B cells, NK cells, and subsets of monocytes) in peripheral blood at days 15, 45, and 70 with respect to baseline

4NCT IDNCT02403505
TitleEarly Phase Clinical Trial About Therapeutic Biological Product Mix for Treating CEA Positive Rectal Cancer
Dates2021/12/28 to 2025/02/28
PhasePhase 1
Enrolment20 (Estimated)
Condition(s)Rectal Cancer
Intervention(s)CEA protein antigen and BCG vaccine mix for percutaneous use
Primary OutcomeTimeframe: up to 90 days
  1. Participants with positive CEA blood test

  2. Participants with positive IGRA blood test with CEA protein antigen after percutaneous use

  3. Participants with IGRA blood test with TB antigens (negative before percutaneous use, positive after percutaneous use)

5NCT IDNCT05507671
TitleThe Role of BCG Vaccine in the Clinical Evolution of COVID-19 and in the Efficacy of Anti-SARS-CoV-2 Vaccines
Dates2021/05/27 to 2023/12/31
PhasePhase 3
Enrolment556 (Estimated)
Condition(s)COVID-19
Intervention(s)BCG vaccine
Primary Outcome
  1. Incidence of SARS-CoV-2 infection. Timeframe: 6 months from recruitment day

  2. Incidence of COVID-19 symptoms. Timeframe: 6 months from recruitment day

  3. Intensity of efficacy of first dose of vaccine against COVID-19. Timeframe: 6 months from recruitment day

  4. Duration of efficacy of the second vaccine dose against COVID-19. Timeframe: 1 year from recruitment day

Selected Secondary Outcomes relevant to Trained ImmunitySerum concentrations of cytokines TNF-α, IFN-γ, IL-1β, IL-4, IL-6, and IL-10 in 50 participants of BCG group versus 50 participants of placebo group 2 months after recruitment
6NCT IDNCT06628544
TitleTrained Immunity in Fungal Infection and Its Mechanism
Dates2020/09/01 to 2023/12/01
PhaseEarly Phase 1
Enrolment79 (Actual)
Condition(s)BCG vaccination
Intervention(s)BCG vaccine
Metformin
Primary OutcomeIL-6 and TNF-α cytokine production by PBMCs isolated after 5 days of continuous medication and restimulated with C. albicans or Mycobacterium tuberculosis
7NCT IDNCT03296423
TitleBacillus Calmette-Guérin Vaccination to Prevent Infections of the Elderly
Dates2017/09/21 to 2020/11/30
PhasePhase 4
Enrollment200 (Actual)
Condition(s)Infection
Hospitalization
Mortality
Intervention(s)BCG vaccine
Primary OutcomeTime to first infection. Timeframe: 12 months
Selected Secondary Outcomes relevant to Trained Immunity
  1. Cytokine stimulation from PBMCs. Timeframe: month 3

  2. Epigenetic changes of circulating monocytes. Timeframe: month 3

8NCT IDNCT02114255
TitleEffects of BCG on Influenza Induced Immune Response
Dates2014/05 to 2014/09
PhasePhases 2 and 3
Enrolment40 (Actual)
Condition(s)Influenza virus infection
Trained Immunity
Intervention(s)BCG vaccine
Primary Outcome
  1. Difference in influenza antibody titers at days 14, 21, 28, and 42

  2. Difference in thrombocyte function at days 0, 14, 21, 28, and 42

Selected Secondary Outcomes relevant to Trained Immunity
  1. IFN-γ, IL-10, type 1 IFN, IL-17, IL-22 production by ex vivo leukocytes stimulated with inactivated/live influenza virus at days 0, 14, 28, and 42

  2. Production of inflammatory mediators (including TNFα, IL-1β, IFN-γ, IL-10, IL-17, and IL-22) by ex vivo leukocytes stimulated with different stimuli (including M. tuberculosis, S. aureus, C. albicans, and inactivated influenza) at days 0, 21, 28, and 42

  3. qPCR/microarray of inflammatory transcriptional pathways at days 0, 14, 21, 28, and 42.

  4. Granzyme B production by ex vivo leukocytes stimulated with inactivated/live influenza virus at days 0, 14, 21, 28, and 42

9NCT IDNCT01734811
TitleEfficacy and Safety Evaluation in Recurrent Wheezing Attacks (MV130)
Dates2012/10 to 2017/02
PhasePhase 3
Enrolment120 (Actual)
Condition(s)Bronchospasm
Bronchiolitis
Bronchitis
Intervention(s)MV130 vaccine
Primary OutcomeNumber of Recurrent Bronchospasm (Wheezing Attacks)
(b) Trials investigating modulation of Trained Immunity for therapeutic benefit
10NCT IDNCT06624436
TitleImmunomodulatory Effects of Dexamethasone, Tocilizumab and Anakinra During Experimental Human Endotoxemia
Dates2024/10/24 to 2025/12
PhasePhase 4
Enrolment52 (Estimated)
Condition(s)Sepsis
Neuroinflammatory Response
Immunosuppression
Endotoxemia
Intervention(s)Dexamethasone
Tocilizumab
Anakinra
Primary Outcome
  1. Plasma TNF concentrations upon second LPS challenge

  2. Cerebrospinal fluid TNF concentrations during repeated experimental human endotoxemia

Selected Secondary Outcomes relevant to Trained Immunity
  1. Plasma cytokine (IL1RA, IL-6, IL-8, IL-10, MIP-1α, MIP-1β, MCP-1, G-CSF, IP-10, CX3CL1, YKL-40) concentrations (plasma and cerebrospinal fluid), other inflammatory protein biomarkers (Olink Target 96 inflammation panel) (plasma and cerebrospinal fluid), and mHLA-DR during first and second LPS challenges

  2. Blood leukocyte single-cell and bulk mRNA profiles/transcriptomic pathways upon LPS challenges

  3. Cytokine production of ex vivo leukocyte cultures

11NCT IDNCT03332225
TitleA Trial of Validation and Restoration of Immune Dysfunction in Severe Infections and Sepsis
Dates2017/12/15 to 2019/12/31
PhasePhase 2
Enrolment36 (Actual)
Condition(s)Sepsis
Macrophage Activation Syndrome
Intervention(s)Anakinra
Recombinant human IFN-γ
Primary OutcomeMortality. Timeframe: 28 days
Selected Secondary Outcomes relevant to Trained Immunity
  1. Cytokine stimulation from PBMCs. Timeframe: 4 and 7 days

  2. Gene expression of PBMCs. Timeframe: 7 days

  3. Epigenetic changes of circulating monocytes. Timeframe: 7 days

(c) Trials investigating inhibition of Trained Immunity for therapeutic benefit
12NCT IDNCT05790499
TitleLDL-c Level Variability and Trained Immunity
Dates2023/03/20 to 2024/01/31
PhaseN/A
Enrollment12 (Estimated)
Condition(s)Cholesterol Variability
Trained Immunity
Intervention(s)Atorvastatin
Primary OutcomeChanges in LDL-C levels between baseline and atorvastatin treatment cycles. Timeframe: 16 weeks
Selected Secondary Outcomes relevant to Trained ImmunityTimeframe: 16 weeks
  1. PBMCs subgroup percentage and activation status

  2. PBMCs secreting cytokines

  3. PBMCs change in gene expression

  4. Levels of hs-CRP, IL-6, IL-18, and sVCAM-1

13NCT IDNCT05210725
TitleTrained Immunity by Dual-pathway Inhibition in Coronary Artery Disease
Dates2022/03/01 to 2022/07/01
PhasePhase 4
Enrolment20 (Actual)
Condition(s)Coronary Artery Disease
Intervention(s)Rivaroxaban and Acetylsalicylic acid
Primary OutcomeWhole blood immune responsiveness to LPS stimulation when switching from acetylsalicylic acid monotherapy to acetylsalicylic acid and low-dose rivaroxaban dual pathway inhibition. Timeframe: 12 weeks
Selected trial outcomes relevant to Trained Immunity
  1. White blood cell count and distribution. Timeframe: 3 months

  2. Monocyte immune responsiveness to LPS stimulation. Timeframe: 3 months

  3. Enrichment of epigenetic gene marks. Timeframe: 3 months

  1. Table of examples of interventional clinical trials related to Trained Immunity. Primary and secondary outcome fields have been simplified from the original data. Only secondary outcomes related to Trained Immunity are included. Source: https://clinicaltrials.gov/.

Appendix 1—table 1
Sources of cells that are potentially suitable for use in models of Trained Immunity.
CellsOriginSpeciesProductSource
MONOCYTES
Stem cellsDYS0100 Induced; Pluripotent Stem CellHumanATCC Product Code: ATCC-ACS-7030https://www.lgcstandards.com/GB/en/search?text=monocyte%20cell%20lines
Cell lineJ774A.1; Monocyte-macrophageMouseATCC Product Code: ATCC-TIB-67https://www.lgcstandards.com/GB/en/search?text=monocyte%20cell%20lines
PrimaryPeripheral Blood CD14+ Monocytes (PBMC)HumanATCC Product Code: ATCC-PCS-800–010https://www.lgcstandards.com/GB/en/search?text=monocyte%20cell%20lines
Cell lineTHP-1; Acute Monocytic LeukemiaHumanATCC Product Code: ATCC-TIB-202https://www.lgcstandards.com/GB/en/search?text=monocyte%20cell%20lines
Cell lineRAW 264.7 gamma NO (-); MacrophageMouseATCC Product Code: ATCC-CRL-2278https://www.lgcstandards.com/GB/en/search?text=monocyte%20cell%20lines
Cell lineU-937; Histiocytic LymphomaHumanATCC Product Code: ATCC-CRL-1593.2https://www.lgcstandards.com/GB/en/search?text=u937
NATURAL KILLER CELLS
Cell lineNK-92; Natural Killer CellHumanATCC Product Code: ATCC-CRL-2407https://www.lgcstandards.com/GB/en/search?text=natural%20killer%20cells
Cell lineNK101HumanRRID:CVCL_WN95https://www.cellosaurus.org/CVCL_WN95
PrimaryCD56+ Natural Killer CellsHumanATCC Product Code: ATCC-PCS-800-019https://www.lgcstandards.com/GB/en/search?text=natural%20killer%20cells
MAST CELLS
PrimaryCD34+ blood cellsHumanRådinger et al., 2010
Cell lineLAD2HumanABM Cat. No.: T8157https://www.abmgood.com/Human-Mast-Cell-Line-LAD2-t8157.html
Cell lineHMC1HumanABM Cat. No.: T8389https://www.abmgood.com/catalog/products/56515/type/cells
Cell lineLUVAHumanABM Cat. No.: T0843https://www.abmgood.com/immortalized-human-mast-cells-luva.html
Cell lineROSA KIT WTHumanRRID:CVCL_5G49https://www.cellosaurus.org/CVCL_5G49
Cell lineROSA KIT D816VHumanRRID:CVCL_5G50https://www.cellosaurus.org/CVCL_5G50
BASOPHILS
Cell lineKU812HumanATCChttps://www.atcc.org/search#q=mast%20cell&sort=relevancy&numberOfResults=24
Bone marrowImmortalizedHumanABM Cat. No.: T0527https://www.abmgood.com/immortalized-human-bone-marrow-basophils.html
NEUTROPHILS
Cell lineHL-60HumanABM Cat. No.: T8970https://www.abmgood.com/hl-60-cells.html
Cell linePLB-985HumanRRID:CVCL_2162https://www.cellosaurus.org/CVCL_2162
Cell lineNB4HumanRRID:CVCL_0005https://www.cellosaurus.org/CVCL_0005
Cell lineKasumi-1HumanRRID:CVCL_0589https://www.cellosaurus.org/CVCL_0589
PrimaryiPSCHumanACS-3002https://www.atcc.org/products/acs-3002
DENDRITIC CELLS
Cell lineBDCMHumanRRID:CVCL_4613https://www.cellosaurus.org/CVCL_4613
Cell lineCAL-1HumanRRID:CVCL_5G46https://www.cellosaurus.org/CVCL_5G46
Cell lineCS-1HumanRRID:CVCL_T022https://www.cellosaurus.org/CVCL_T022
Cell lineFDC-1HumanRRID:CVCL_0F06https://www.cellosaurus.org/CVCL_0F06
Cell lineMUTZ-3HumanRRID:CVCL_1433https://www.cellosaurus.org/CVCL_1433
EOSINOPHILS
Cell lineAML14HumanRRID:CVCL_8286https://www.cellosaurus.org/CVCL_8286
Cell lineEOL-1HumanRRID:CVCL_0258https://www.cellosaurus.org/CVCL_0258
iPSCHPS1472HumanRRID:CVCL_UN83https://www.cellosaurus.org/CVCL_UN83
NONIMMUNE CELLS
Cell linesFibroblasts20,869 Cell lineshttps://www.cellosaurus.org/search?query=fibroblast
Liu et al., 2016
Epithelial cells5444 Cell lineshttps://www.cellosaurus.org/search?query=epithelial
Naik et al., 2017
  1. The cell lines listed in this table are examples only and form a subset of all of the available cell lines.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Jelmer H van Puffelen
  2. Callum Campbell
  3. Irene Gander-Meisterernst
  4. Johanna Holldack
  5. Pauline T Lukey
(2025)
Trained Immunity: RoadMap for drug discovery and development
eLife 14:e108465.
https://doi.org/10.7554/eLife.108465