imperial college london · web viewcellular immunotherapy cellular therapy represents a promising...

45
Immunotherapeutic approaches to fungal diseases: current state of the art and future perspectives Darius Armstrong-James MD 1* , Gordon D. Brown PhD 2 , Mihai G. Netea MD 3 , Teresa Zelante PhD 4 , Mark S. Gresnigt PhD 3 , Frank L. van de Veerdonk MD 3 , Stuart M. Levitz MD 5 1 * Corresponding author. Fungal Pathogens Laboratory, National Heart and Lung Institute, Imperial College London, UK SW7 6NP email: [email protected] Tel: +4420 7594 2746 2 Aberdeen Fungal Group, MRC Centre for Medical Mycology, University of Aberdeen, Aberdeen, UK, AB25 2ZD 3 Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands 4 Department of Experimental Medicine, University of Perugia, 06132 Perugia 5 Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01665

Upload: others

Post on 25-Feb-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Immunotherapeutic approaches to fungal diseases: current state of the art and future

perspectives

Darius Armstrong-James MD1*, Gordon D. Brown PhD2, Mihai G. Netea MD3, Teresa Zelante

PhD4, Mark S. Gresnigt PhD3, Frank L. van de Veerdonk MD3, Stuart M. Levitz MD5

1* Corresponding author. Fungal Pathogens Laboratory, National Heart and Lung Institute,

Imperial College London, UK SW7 6NP

email: [email protected]

Tel: +4420 7594 2746

2Aberdeen Fungal Group, MRC Centre for Medical Mycology, University of Aberdeen,

Aberdeen, UK, AB25 2ZD

3Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud

University Medical Center, 6525GA Nijmegen, The Netherlands

4Department of Experimental Medicine, University of Perugia, 06132 Perugia

5Department of Medicine, University of Massachusetts Medical School, Worcester,

Massachusetts 01665

Page 2: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Abstract

Fungal infections cause significant morbidity worldwide and are associated with an

unacceptable high mortality despite the availability of antifungal drugs. The incidence of

mycoses is rising due to the HIV pandemic, coupled with the increasing use of

immunomodulatory medical interventions that are used to treat autoimmune diseases and

cancer. Treatment with new classes of antifungal drugs has only resulted in partial success in

improving the prognosis of patients with fungal infection. Therefore, adjunctive host

directed therapy is believed to be the only option to further improve the outcome of these

patients. Recent understanding in the complex interaction between fungi and the host has

led to the design and exploration of novel therapeutic strategies such as cytokine therapy,

vaccine development, and cellular immunotherapy, which might prove to be viable adjuncts

to our current antifungal regimens. We will discuss the rationale, challenges, and progress

of these immunotherapeutic approaches that are so urgently needed to overcome the

devastating impact of fungal diseases.

Page 3: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Introduction

Fungal infections occur in more than a quarter of the world’s population, yet their

enormous influence on human health is not widely appreciated. The majority of these

infections are superficial, and most are easily treatable. However, fungi also cause invasive

diseases with mortality rates exceeding 50%, and which are estimated to result in more than

one and a half million deaths every year on a global scale.1,2 These unacceptably high

mortality rates stem primarily from inadequacies in the current fungal diagnostics, and the

clinical shortcomings of currently available antifungal drugs. The majority of invasive disease

is caused by only four genera of fungi, Candida, Aspergillus, Cryptococcus and Pneumocystis,

yet there are no clinical vaccines currently for these fungi1.

Most invasive infections occur as a result of altered immune status. The increased

incidence of these diseases has arisen primarily as a result of greater use of

immunosuppressive and invasive medical interventions, and the HIV pandemic. The key role

of defective host immunity in fungal disease has led to interest in developing adjunctive

immunotherapies that can enhance critical immune effector functions and improve

outcomes. However, targeted immunotherapies require comprehensive insight into the

host response against fungi. Exciting recent advances in our understanding of the

mechanisms of protective anti-fungal immunity have provided a fundamental basis for the

development of these new approaches. We now appreciate the central role of phagocytic

cells in protective innate host responses and in controlling the development of adaptive

antifungal immunity.3,4 For example, neutropenia or defects in phagocyte NADPH oxidase

(chronic granulomatous disease, CGD) lead to life-threatening fungal infections.3 The

Page 4: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

activities of phagocytes present therapeutic opportunities, as they can be enhanced or

suppressed by soluble immunomodulatory mediators. We now also realize the importance

of pattern recognition receptors, particularly the C-type lectin receptors, and their signalling

pathways, in antifungal defence.5 For example, polymorphisms in the Toll-like receptors and

C-type lectin receptors, have been linked to susceptibility to various fungal infections in

patients.6 Knowledge of these receptor systems and their cognate fungal ligands also offers

opportunities for the development of adjuvants and vaccines.

Substantial advances have also been made in our understanding of roles of pattern

recognition receptors, and the leukocytes they are expressed on, in the modulation of

adaptive immunity. For example, interactions of fungi with C-type lectin receptors, such as

Dectin-1, Dectin-2, mannose receptor, or Mincle, induce both Th1 and Th17 immunity.4

While experimental studies report the effects of NK and other innate lymphocytes on

outcome of fungal infections, more studies are needed to assess their role in clinical disease.

Progress in our understanding of these systems and their underlying mechanisms also

provide considerable opportunities for the development of novel immunotherapies, such as

inducing protective CD8+ responses in AIDS patients or by treating cryptococcal meningitis

with adjunctive interferon-γ (IFNγ) immunotherapy.7,8

Despite these substantial advances, our knowledge is limited largely to immune

responses to the four major fungal pathogens listed above. We have much less knowledge

about immunity to most other fungal pathogens, including the highly prevalent fungal

infections that cause superficial, non-life-threatening diseases. In addition, knowledge on

the immunity to rare fungal pathogens such as the Mucorales is also scarce, despite

extremely high mortality. We are only just beginning to appreciate the importance of fungi

Page 5: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

to immune homeostasis, and the pathology of inflammatory and autoimmune diseases,

such as colitis.9

Nevertheless, novel host directed therapies have an enormous potential to change

the future of antifungal treatment. In this review, we discuss the recent progress in

immunotherapeutic antifungal approaches including cytokine therapy, vaccine

development, and cellular immunotherapy.

Recombinant cytokines as adjunctive therapy in systemic infections

The use of recombinant cytokines and growth factors represents an important

strategy for host-directed therapy in fungal infections. Recent advances in our

understanding of the genetic causes of fungal infections provide an important source of

information: by understanding which immune defects lead to increased susceptibility to

fungal infections, appropriate adjunctive therapy can be designed. A large number of

primary immunodeficiencies have been shown to increase susceptibility to fungal infections,

as reviewed in detail.10,11 In Table 1 we show the major genetic causes of increased

susceptibility to fungal infections, which emphasize the critical function of neutrophils and

T-helper lymphocytes for effective antifungal host defences. These conclusions are also

strongly supported by information from patients with acquired neutrophil (chemotherapy,

immunosuppressive therapy) or CD4 T lymphocyte (HIV, immunosuppressive therapy)

immunodeficiencies who are highly susceptible to fungal infections. The importance of

neutrophils in the host defence against Candida and Aspergillus infection, and the

importance of Th1 and Th17 adaptive responses, provided guidance for developing

Page 6: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

strategies to augment host defence against fungi, with colony-stimulating factors (CSFs) and

recombinant IFNγ (rIFNγ) as the main classes of biologicals that are available to enhance

antifungal host defences.

CSFs were among the first candidate antifungal adjunctive therapies, due to their

dual effect on myeloid cell population recovery, and activation of neutrophils. Granulocyte

macrophage-CSF (GM-CSF) stimulates expression of the C-type lectin, Dectin-1, on

macrophages and ameliorates the course of disseminated murine candidiasis.12 Adjunctive

immunotherapy for refractory mucosal candidiasis in HIV-positive patients was successful

and without adverse events.13,14 While no studies with GM-CSF have been performed in

patients with invasive candidiasis specifically, either GM-CSF or GM-CSF and G-CSF

combination therapy is associated with a reduced post-transplant mortality, and with

reduced incidence of Candida infections.15 A combination of treatment with rGM-CSF and

rIFNγ reportedly induced immunological and clinical recovery in three patients with invasive

aspergillosis.16 Although no systematic studies to date have assessed the clinical efficacy of

GM-CSF in cryptococcosis, this is a rational choice given the increase in susceptibility to

Cryptococcus in patients with neutralizing anti-GM-CSF antibodies.17

Recombinant granulocyte-CSF (rG-CSF) also increases neutrophil numbers and

enhances their antifungal activity, and is routinely used to shorten neutropenia during

oncologic treatments. In a randomized placebo-controlled pilot study in non-neutropenic

patients with disseminated candidiasis, adjunctive immunotherapy with rG-CSF and

fluconazole was showed a trend towards faster resolution of infection compared to

fluconazole alone.18 Nevertheless, a note of caution has to be given as rapid neutrophil

recovery on G-CSF therapy can lead to severe pulmonary complications,19 illustrating the

Page 7: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

delicate balance between restoration of antifungal resistance and induction of pathology.

The cytokine that has been best studied as an antifungal adjuvant immunotherapy is

rIFNγ. rIFN can be used as repletion therapy in patients with Th1 synthesis defects (e.g.,

patients with STAT3 or IL-12R deficiencies), as well as in a pharmacological fashion leading

to supraphysiological concentrations (e.g. CGD patients). rIFNγ enhances antifungal activity

of macrophages and PMN, and rIFNγ immunotherapy reduces fungal burden in murine

candidiasis,20 aspergillosis 21 and cryptococcosis.22 The first successful clinical applications of

rIFNγ as an adjunctive therapy was in CGD, providing significant protection against

aspergillosis.23,24 rIFNγ treatment may also have a place in transplantation patients with

systemic fungal infections, where effective clearance of refractory or disseminated fungal

infections (especially aspergillosis) was shown in several patients.25 These studies are the

basis for the use of rIFNγ immunotherapy in patients with aspergillosis; however controlled

randomized trials are needed.

IFNγ concentrations at the site of infection determine the rate of cryptococcal

clearance,26 and a clinical trial of adjuvant immunotherapy with rIFNγ in HIV-positive

patients with cryptococcal meningitis showed faster sterilisation when compared to

standard therapy alone.7 In a second trial, adjunctive short-course rIFNγ significantly

increased CSF cryptococcal clearance without increases in adverse events.27 The efficacy of

IFNγ therapy has also been reported in CD4 lymphopenia 28 suggesting that IFNγ

immunotherapy is an important adjuvant weapon for the treatment of cryptococcal

meningitis.

The importance of Th1 responses in Candida infection suggests a potential benefit of

Page 8: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

IFNγ immunotherapy in patients with invasive candidiasis. In an open-label, prospective

pilot study, 6 patients with candidaemia received either adjunctive immunotherapy with

rIFNγ or placebo. Adjunctive immunotherapy with rIFNγ improved leukocyte innate (IL-1,

TNFα) immune responses, increased production of the T-cell cytokines IL-17 and IL-22, and

restored mHLA-DR expression.29 Although this study was not powered for mortality, its

results demonstrate the potential for adjunctive immunotherapy with rIFNγ to enhance

antifungal immunity and restore sepsis-associated immunoparalysis in candidemia patients.

Significant beneficial effects on clinical outcome have been observed with the use of IFNγ in

CGD and cryptococcal meningitis27. While an increasing number of studies suggest that this

may be the case in other invasive fungal infections, larger randomized trials are needed.

Immune dysregulation as a risk for fungal infections

Although severe induced and acquired immunodeficiencies strongly impact host

immune defences and greatly increase the risk of invasive fungal infections, human and

mouse studies show that hyperreactivity of both innate and adaptive arms of the immune

response also predispose to invasive fungal infections. For example, the immune

reconstitution inflammatory syndrome (IRIS) is a complication of antiretroviral therapy in

immunocompromised patients with HIV.30 Paradoxically, immunosuppression and

corticosteroids have been used to treat more severe cases of IRIS associated with

concomitant invasive fungal infections; by lowering levels of immune reactivity, the fungal

infection is once again brought under control6. Hence immune system dysregulation can be

considered a primary cause of susceptibility to fungal infections, and opportunistic fungal

Page 9: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

infections reconsidered as immune-mediated pathologies.

These advances in understanding have overturned the belief that successful immunity

relates only to the blockade of infective capacity of the pathogen. Informed by established

theories of evolutionary ecology,31 we can now revise our definition of immunity to

pathogens: a successful immune response is the process by which the host both resists

infective capacity of a microorganism, and limits the damage caused either directly or

indirectly by the infection. In the case of fungal infection, damage-limiting tolerance and

pathogenic resistance have been proposed as two axes that frame the space within which

the immune response occurs; the point of their intersection determines the balance of

these critical factors, and therefore the overall benefit to the host of that response.32,33

Experimental data to refine these theories are in the earliest stages of emerging, however

studies in murine models of candidiasis and aspergillosis have so far identified Th1 cells as

key mediators of antifungal resistance, and Treg cells as most involved in damage-

limitation.34 Bringing this concept to the clinical arena, immune dysregulation implies

alterations of mechanisms that either enhance the immune system’s battle against fungal

microbes or abrogate the tolerance that controls the self-harm caused by an immune

response, known as immunopathology (Figure 1).

There are several lines of evidence that maintaining self-tolerance has equal

importance to providing pathogen resistance in immunity. One situation with immediate

clinical relevance is in patients with primary immunodeficiencies (PIDs), who can exhibit

markedly dysregulated immune responses leading to both susceptibility to infection and

significant autoimmune manifestations. For example, autoimmune polyendocrinopathy-

candidiasis-ectodermal dystrophy (APECED) is a PID that is characterized by simultaneous

Page 10: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

dysregulation of immune tolerance and multiple fungal infections.3511 Candida infections are

very common in patients affected by APECED, where autoantibodies are released against

cytokines involved in both resistance and tolerance arms of the response.36 In these

conditions, chronic mucocutaneous candidiasis is also frequently diagnosed; a clinical

syndrome with selectively altered immune responses against Candida consistent with

immune dysregulation.37 Interestingly, APECED is characterized by mutations in the

autoimmune regulator (AIRE) gene, which is mainly involved in central tolerance induction.

Further studies in non-HIV-infected individuals with cryptococcal meningities show that

severe disease is associated with M@ macrophage polarisation, intrathecal expansion of

activated HLA-DR+ CD4+ and CD8+ T cells and NK cells, high levels of interferon-, IL-4 and

IL-13, and neuronal damage in the face of good microbial control38. These observations give

further evidence for immune dysregulation as a major driver of immunopathology in fungal

disease.

The presence of autoantibodies against IFN- has also been linked with fungal infections.17,39

Accordingly B-cell depleting therapies, such as rituximab, have been successfully tested in

patients suffering from mycobacterial infection,40 however this strategy has yet to be

extended to the fungal infection setting. Similarly to IRIS, chronic disseminated candidiasis

(CDC), also called hepatosplenic candidiasis, is a severe invasive fungal infection principally

observed during neutrophil recovery in patients previously treated with

immunosuppressant therapies. Recent studies have shown that the addition of

corticosteroids to antimicrobial therapy is more effective than antifungal drugs alone in

reducing inflammation, whilst treating fungal infection41. Similarly, it is well established that

PRR polymorphisms predispose towards vulvovaginal candidiasis (VVC)42, but more recently,

Page 11: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

evidence of pathogenic inflammasome activity has also been observed in VVC.43 In murine

experimental candidiasis, a pathogenic inflammatory response characterized by hyper-

active type-1 IFN signaling promotes inflammatory DC differentiation, and thereby fungal

infection44, though whether a parallel mechanism operates in humans remains to be seen.

In addition to autoantibodies against IL-17, deficiency in the subunits of the IL-17 receptor

(IL-17RA and IL-17RC), IL-17F itself, the IL-17R adapter molecule ACT1, and defects in the

STAT1 adaptor molecule (see below) are associated with the development of chronic

mucocutaneous candidiasis, underscoring the importance of the IL-17 signalling pathway in

the pathogenesis of CMC45,46. While some PIDs are characterized by dysregulation of

tolerogenic functions, others present with altered inflammatory/resistance responses, for

example in Th17 development or function, and concomitant occurrence of fungal

infections.. 16As discussed above, mutations in STAT3 (in autosomal dominant Hyper IgE

syndrome), CARD9, Il-17ra and Il-17f, result in dampened Th17 adaptive inflammatory

response and therefore diminished resistance to fungi.471 STAT1 gain of function (GOF)

mutations underlie AD-CMC and are associated with a large spectrum of features ranging

from auto-immune disease to cancer to cerebral aneurysm formation48. Since cytokines,

such as IFN and type 1 IFNs signal via JAK1 and activate STAT1, JAK inhibitors have been

explored in patients with STAT1 GOF mutation. Some of these attempts have been

successful49,50. Given the broad clinical phenotype and the complexity of the underlying

pathophysiology it remains to be investigated whether this approach is beneficial in a subset

of patients with AD-CMC due to STAT1 GOF mutations. PIDs may also incorporate genetic

defects in the primary innate immune response, in particular in phagocytic functions, as in

CGD where there is reduced phagocytic ability, an altered respiratory burst in response to

Page 12: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

pathogens, and a specific defect in LC3-associated phagocytosis, a crucial mechanism for

efficient uptake of Aspergillus51,52 Interestingly, CGD also leads to dysregulated Th17 cell

expansion, with high levels of IL-1 and IL-17 correlated with susceptibility to

aspergillosis.53,5418,19 Ultimately, immunotherapies, such as blockade of IL-1 with Anakinra

might prove to be an important adjunctive strategy in the treatment or prevention of fungal

infections in CGD, where the critical aim is to re-establish the balance between immune

resistance and immune tolerance.52

Immunomodulation in fungal allergic diseases

An exaggerated immune response to fungi may cause allergic diseases ranging from

atopic dermatitis and asthma with fungal sensitisation to allergic bronchopulmonary

aspergillosis (ABPA), a severe allergic pulmonary complications caused by Aspergillus spp.

Pathological inflammatory responses in ABPA are mainly characterized by excessive Th2

responses resulting in uncontrollable asthma, mucosal plugging and bronchiectasis,

sometimes leading to fatal lung damage. The current treatment for ABPA is oral

corticosteroids, control of the underlying asthma with β2-agonists, and azole antifungal

treatment.55,56 Due to the potential severe side effects of azoles and corticosteroids, new

immunotherapies for ABPA are urgently needed, for which a better understanding of the

underlying immunological mechanisms will be crucial.

Allergic reactions can be triggered by fungal cell wall components that stimulate the

production of thymic stromal lymphopoietin, subsequently inducing chemokines such as

TARC (CCL17) which have been shown to play an important role in both murine and human

Page 13: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

asthma with fungal sensitisation.57-59 TARC binds to its receptor CCR4 and induces a Th2

response that drives mast cell degranulation and increases pulmonary IgE levels. 60 The

hallmark of allergic inflammatory responses is increased production of the Th2 cytokines IL-

4, IL-5 and IL-13 and the production of IgE. OX40L is a co-stimulatory molecule on antigen

presenting cells that can potentiate these Th2 responses.61 Aspergillus-induced Th2

responses in vivo were dependent on OX40L that in turn was dependent on vitamin D in

experimental murine fungal asthma.61 Vitamin D decreased OX40L expression, reduced Th2

responses and was therefore tested recently in a clinical trial in patients with cystic fibrosis

(CF) and ABPA.62 This phase-1 trial with vitamin D reduced the Aspergillus-induced IL-13 and

IgE responses in patients with ABPA and was well tolerated.62 Another approach to limiting

Th2 responses is to target IgE with the monoclonal antibody omalizumab. Although not

approved, omalizumab has been successfully used in ABPA with and without CF allowing a

corticosteroid-sparing treatment regimen.63-66

Although much work in the field of asthma has not addressed the fungal allergy

component in detail, there is a spectrum ranging from neutrophilic (Th1/Th17) to

eosinophilic asthma (Th2), suggesting that asthma with fungal sensitisation might also have

a neutrophilic component. Peripheral blood mononuclear cells isolated from patients with

ABPA have decreased production of IFNγ in response to Aspergillus and exaggerated Th2

responses.67 This increased Th2/Th1 ratio was also observed in asthma patients with

Aspergillus-sensitization, but not in patients with asthma without Aspergillus-sensitization.67

These findings provide a rationale to explore immunomodulatory treatment with rIFNγ in

Aspergillus-driven allergic inflammatory disorders. Other studies suggest IL-33 as a

therapeutic target in severe asthma with fungal sensitization. 68

Page 14: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Experimental Aspergillus-induced airway inflammation can have different

characteristics depending on the mouse strain examined. BALB/c mice display increased

neutrophilia in the lung and more potent TNFα responses compared to C57BL/6 mice.69

When the pulmonary dendritic cells responsible for the TNFα production in BALB/c mice

were depleted, the inflammatory responses switched from a TNFα-IL-17 signature with

predominant neutrophilic infiltrate to an increased IL-5 response with increased eosinophil

influx resembling the inflammatory response observed in C57BL/6 mice.69 Thus fungi can

trigger different types of pulmonary responses depending on the host genetic background.

Host directed therapy in fungal allergy diseases can therefore be based on several

strategies: (i) dampening neutrophilic inflammation (e.g., by blocking TNFα, IL-1 or IL-17); (ii)

inhibiting Th2 responses (blocking IgE or IL-13); or (iii) augmenting antifungal immune

responses (IFNγ or GM-CSF) (Figure 2).

Fungal vaccines and antibody therapy

There have been considerable efforts to develop preventive vaccines and

immunotherapies for fungal disease; however it is worth noting some of the obstacles that

have impeded vaccine development. Fungi are ubiquitous in the environment; the rarity of

direct person-to-person spread implies that fungal vaccines cannot be expected to impart

herd immunity on a population. Furthermore most individuals with serious fungal infections

are immunocompromised and likely to mount suboptimal responses to vaccination.70

Nevertheless, vaccination could be considered in individuals with relatively intact immunity

anticipated to have severe immunosuppression in the future. Examples include persons

awaiting solid organ transplant and those with early HIV infection. Another attractive target

for vaccination is people living in or traveling to areas endemic for Coccidioides and

Page 15: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Histoplasma. Finally, efforts are underway to develop vaccines that downregulate the

allergic response in persons with asthma with fungal sensitization.

An attractive fungal vaccine strategy targets shared antigens of the most common

genera of medically important fungi. For example, a key component of the cell wall of fungi

is β-1,3-D-glucan; but this glycan is poorly immunogenic. Nevertheless, mice immunized

with the β-1,3-D-glucan, laminarin, conjugated to diphtheria toxoid develop strong antibody

responses and are protected in models of candidiasis, aspergillosis and cryptococcosis.71

Furthermore, immunization of mice with antigens encapsulated within glucan particles

results in long-lasting and protective antigen-specific antibody and T cell responses.72

Promising results have been obtained in preclinical studies of vaccines composed

live, attenuated fungi.(reviewed in 73) Vaccination with an attenuated strain of the endemic

fungus, Blastomyces dermatitidis, protected mice against subsequent challenge with a

virulent strain.8 Remarkably, protection was seen even in the setting of CD4+ T cell depletion

due to the emergence of protective CD8+ T cells. Another live vaccine strategy that

successfully protected CD4+ T cell deficient mice used a C. neoformans strain engineered to

produce murine IFNγ.74 Such studies have obvious implications for the development of

vaccines that protect in the setting of AIDS and other CD4+ T cell deficiencies.

A caveat to the use of live vaccines is they need to be sufficiently attenuated to not

cause disease, particularly in immunosuppressed hosts. Killed whole-cell vaccines have

shown promise in murine models of fungal infections.73 In a phase-3 clinical trial, recipients

of a vaccine composed of formaldehyde-killed spherules of Coccidioides immitis had a 25%

prevention rate against coccidioidomycosis compared to control subjects, which was non-

Page 16: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

significant.75 While these studies demonstrated the feasibility of conducting human fungal

vaccine trials, they also highlighted that the proinflammatory properties of fungi can elicit

unacceptable local inflammatory reactions. For this reason, much of the emphasis in

modern fungal vaccine development has been on subunit vaccines. Two vaccines that

incorporate recombinant C. albicans-derived proteins demonstrated immunogenicity in

Phase-1/2 clinical trials.76,77

The recognition that antibodies elicited by vaccination could protect against fungal

infection has led investigators to produce therapeutic monoclonal antibodies. In mouse

models, the protective efficacy of monoclonal antibodies against cell wall β-1,3-D-glucan

and C. neoformans capsule has been demonstrated. Interestingly, in addition to

opsonization, antifungal effects of antibodies have been attributed to direct inhibitory

effects on fungal growth and metabolism.71,78 Administration of a monoclonal anticapsular

antibody to persons with AIDS and cryptococcal meningitis resulted in a transient reduction

in serum cryptococcal antigen titers.79

Cellular immunotherapy

Cellular therapy represents a promising approach to treating infection, by redirecting

the power of immune cells through intelligent design to attack foreign invaders. In the 1960s

adoptive cell transfer (syngeneic or allogeneic transfer of cells into an individual) emerged

as an exciting approach for the treatment of malignancies with T cells.80 Major progress in

the field was made with the discovery that immunoglobulin T cell receptor chimeric

molecules could be expressed, heralding the advent of designer T cells that can be directed

to kill based on specific molecular targets.81 Whilst T cell-based therapy holds immense

Page 17: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

potential, there are significant risks associated with the potential for cytokine storms. 82 In

parallel, innate cell therapy has steadily gathered pace as a promising alternative approach,

since the initial use of granulocyte transfusions for neutropenic sepsis, with the advantage

that there is no need to re-engineer antigen specificity.83

Adoptive T cell therapy and chimeric antigen receptor T cell engineering

The rationale for adoptive T cell therapy for fungal disease is the observation that anti-

Aspergillus T cells are slow to engraft after allogeneic haematopoetic stem cell

transplantation.84 An early clinical study showed that adoptive transfer of anti-Aspergillus T

cells led to enhanced control of Aspergillus antigenaemia, and reduced mortality.85 Further

studies refining selection and expansion protocols for anti-Aspergillus T cells, have primarily

focused on optimal antigen-based selection protocols.86 Recent studies have raised the

potential of selection of multi-pathogen T cells against the major medically important

fungi.87 A key question is around whether or not specific antigen-based selection or broad

repertoire selection using fungal extracts is optimal, with further unanswered questions

around the relative utilities of CD4 and CD8 T cells, and the ability of specific antigens to

induce specific Th1, Th2 or Th17 cells. One possible drawback of T cell therapy is the

potential for alloreactivity with the recipient, however studies thus far indicate this is

unlikely to be a major problem.85

Re-engineering of T cells with chimeric antigen receptors have shown huge potential

for the treatment of individuals with B cell malignancies.88 Fundamentally, chimeric antigen

receptor (CAR) technology consists of a extracellular ligand recognition domain, typically a

single-chain variable fragment, to a intracellular signalling complex including CD3ζ to enable

Page 18: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

T cell activation after antigen binding.88 Enhanced activation can be achieved by adoption of

further co-stimulatory molecules such as CD28, 4-1BB, or OX40. Adapting such an approach,

Kumaresan and colleagues recently showed that CAR technology can be modified for fungal

disease, by substituting the variable fragment for the fungal-specific C-type lectin receptor

Dectin-1 (Figure 3).89 Resulting D-CAR T cells exhibited specificity for β-1,3-D-glucan and

were effective against A. fumigatus in vitro and in vivo in murine models. This pivotal study

demonstrates proof-of principle that that CAR technology can be extended to encompass

CTLs, critical innate pattern recognition receptors with broad repertoires against fungal

pathogens.

Innate cellular therapy

The association between neutropaenia and severe sepsis has been long recognised,

particularly in haematology medicine. Granulocyte transfusion emerged as a rational

approach in the 1970s, but was hindered by problems around toxicity and superceded by

the advent of multiple broad-spectrum antimicrobial agents and growth factors in the

1980s. However, there has been renewed interest in granulocyte transfusion, especially

with the availability of recombinant G-CSF and GM-CSF that are used to greatly increase the

yield of granulocytes from donors.83 Two contemporary randomised studies in febrile

neutropaenia have been undertaken. The first study recruited 55 patients with fungal

disease. There was no obvious difference in survival, however the study was beset by

logistical difficulties such as delays in granulocyte transfusions, and closed early.90 A more

recent multicentre randomised study showed no difference in overall success compared to

antibiotics alone, however those individuals who received higher doses of granulocytes

tended to have better outcomes.91 Again, the study failed to meet enrolment targets.

Page 19: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Most work with dendritic cells for fungal immunotherapy is pre-clinical with promising

studies in murine transplantation models.92 Priming of dendritic cells with either Aspergillus

conidia or fungal RNA led to the subsequent activation of fungal-specific Th1 T cells and

enhanced protection from invasive aspergillosis. NK cells hold the advantage that they do

not cause graft-versus-host disease during adoptive transfer. In addition, they are able to

damage the hyphae of A. fumigatus and Rhizopus arrhizus through the release of perforin,

and play a crucial role in models of neutropaenic aspergillosis.93,94 As clinical trials are

already under way to assess NK cell adoptive therapy in malignancies, future studies to

address their utility in fungal disease are a new goal for the field.95 Other approaches include

innate stimulation, such as with imiquimod in chromoblastomycosis (a chronic skin

infection), where deficiencies in Toll-like receptors receptor recognition were shown to

underlie disease susceptibility.96 97

Immunotherapy and risk stratification of patients

In recent years, numerous studies have emerged that demonstrate association of common

genetic polymorphisms with increased risk for fungal infections; especially in at risk

populations such as transplant patients. (Reviewed in 98,99). It may be possible to stratify

patient risk of infection based on immunogenetics. Such patients would benefit from more

intensive diagnostic screening or prophylactic antifungal therapy, and by knowing the exact

defective immune pathway targeted immunotherapy that circumvents the defect can be

prescribed.

The fact that immunotherapies are targeted towards specific immune defects or

dysregulation of the antifungal host response highlights the importance of characterising

Page 20: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

the host’s immune status. With progression of the infection and the introduction of immune

therapy the antifungal host response can change over time, and the immunotherapy might

not be required anymore or needs to be adjusted. New functional immunoassays are

therefore needed that can define host deficits and how they respond to immunotherapy.

100,101

Conclusions

Fungal diseases continue to cause devastating high mortality infections in the context

of primary and acquired immunodeficiencies globally. The close relationship to

immunocompromised status, combined with poor outcomes and increasing resistance to

conventional antifungal chemotherapy, has pushed immunotherapy to the fore. The current

rapid progress in the clinical immunotherapy field as a whole presents unprecedented

opportunities to exploit current approaches for fungal disease, from recombinant cytokines,

to vaccines, monoclonal antibodies, and designer T cells. Whilst substantial laboratory-

based progress has been made, and some important clinical studies conducted, the major

challenge for the next decade will be to drive forward clinical trials that conclusively show

the utility of immunotherapy for fungal diseases.

Contributors

All authors contributed to the writing of this manuscript. MSG and TZ prepared the figures.

Conflict of interest

We declare that we have no conflicts of interest.

Page 21: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Search strategy and selection criteria

The PubMed database was searched using the following searching criteria: the terms “fungal ” and “immunotherapy”. From the search result the most relevant primary articles were selected. Only articles in English were reviewed. Relevant references included in these publications were also used.

Page 22: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Table 1. Some of the most important genetic causes of severe fungal infections.

Fungal infection Genetic cause Mechanism

Systemic/CNS Candida CGD, CARD9-deficiency,

MPO deficiency

Defective ROS production,

defective Th17/neutrophil

recruitment

Chronic mucosal

candidiasis

STAT1-GOF, STAT3-

deficiency, AIRE-deficiency

(APECED), IL17F/IL-17RA

deficiency, ACT1 deficiency,

IL17RC deficiency, DOCK8

deficiency

Defective Th17,

autoantiobodies against

IL-17/IL-22

Aspergillosis CGD, STAT3-deficiency,

GATA2 deficiency

Defective ROS production,

defective Th17/neutrophil

recruitment, defective

haematopoiesis

Deep dermatophytosis CARD9-deficiency, STAT1-

GOF

defective Th17/neutrophil

recruitment

Cryptococcosis Autoantibodies against GM-

CSF or IFNγ, primary CD4

lymphopenia, GATA2

deficiency

Neutralization of GM-CSF/IFNγ

bioactivity, defective Th

function, defective

haematopoiesis

Dimorphic fungi Interleukin-12Rb1

deficiency, IFNγR deficiency,

STAT1-GOF, GATA2

deficiency

Defective Th1 function,

defective Th17 function,

defective haematopoiesis

Pneumocystosis SCID, X-linked CD40 ligand

deficiency, CARD11

deficiency, MHC class II

deficiency, NEMO

deficiency

Defective CD4 lymphocyte

function, defective NFkB

modulation

Page 23: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Abbreviations: dimorphic fungi (histoplasmosis, coccidioidomycosis,

paracoccidioidomycosis), APECED (autoimmune polyendocrinopathy-candidiasis-ectodermal

dystrophy), CGD (chronic granulomatous disease), Th (T-helper lymphocyte)

Page 24: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

Figure legends

Figure 1. The yin and yang of Immunomodulation in fungal diseases.

Substantial advances now indicate that immune responses to fungal infections can polarize

to either ‘resistance’ or ‘tolerance’ arms of defence, promoting or inhibiting antifungal

immune reactions, respectively. From the concerted and balanced action of the two

immune arms, mammalian host fitness to fungi is achieved, raising the question whether

immunotherapy in human fungal diseases should aim for the same duality.

Figure 2. Immunomodulatory options in Aspergillus -induced pulmonary allergic diseases.

Aspergillus hyphae can trigger excessive Th2 responses. Host-directed therapy with the anti-

IgE monoclonal antibody omalizumab, and blocking OX40L expression on antigen presenting

cells with vitamin D might decrease exaggerated Th2 responses induced by Aspergillus. IFNγ

could supplement the deficient IFNγ response in ABPA, increase Aspergillus clearance via

stimulation of phagocytosis and killing, and dampen Th2 responses. Furthermore, when a

predominant neutrophilic signature is present, blocking cytokines such as TNFα, IL-1 and IL-

17 with biologicals such as anti-TNFα, anakinra, and anti-IL-17 might be beneficial.

Figure 3. Designer T-cells for fungal therapy: molecular anatomy of a D-CAR T cell.

Re-engineering T cells with specificity for fungal pathogens through the exploitation of C-

type lectin receptor function represents an exciting and novel approach for fungal disease

Page 25: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

therapy. In proof of principle studies the classic C-type lectin fungal receptor Dectin-1

extracellular region was expressed in combination with CD28 and CD3 cytoplasmic regions

to enable -(1,3)-D-glucan-specific activation of T cells in response to A. fumigatus.

Page 26: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

References

1. Brown GD, Denning DW, Gow NA, Levitz SM, Netea MG, White TC. Hidden killers: human fungal infections. Sci Transl Med 2012; 4(165): 165rv13.2. Vallabhaneni S, Mody RK, Walker T, Chiller T. The Global Burden of Fungal Diseases. Infect Dis Clin North Am 2016; 30(1): 1-11.3. Brown GD. Innate antifungal immunity: the key role of phagocytes. Annual review of immunology 2011; 29: 1-21.4. Wuthrich M, Deepe GS, Jr., Klein B. Adaptive immunity to fungi. Annu Rev Immunol 2012; 30: 115-48.5. Hardison SE, Brown GD. C-type lectin receptors orchestrate antifungal immunity. Nature immunology 2012; 13(9): 817-22.6. Carvalho A, Cunha C, Pasqualotto AC, Pitzurra L, Denning DW, Romani L. Genetic variability of innate immunity impacts human susceptibility to fungal diseases. Int J Infect Dis 2010; 14(6): e460-8.7. Pappas PG, Bustamante B, Ticona E, et al. Recombinant interferon- gamma 1b as adjunctive therapy for AIDS-related acute cryptococcal meningitis. J Infect Dis 2004; 189(12): 2185-91.8. Wuthrich M, Filutowicz HI, Warner T, Deepe GS, Jr., Klein BS. Vaccine immunity to pathogenic fungi overcomes the requirement for CD4 help in exogenous antigen presentation to CD8+ T cells: implications for vaccine development in immune-deficient hosts. J Exp Med 2003; 197(11): 1405-16.9. Iliev ID, Funari VA, Taylor KD, et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 2012; 336(6086): 1314-7.10. Plantinga TS, Johnson MD, Scott WK, et al. Human genetic susceptibility to Candida infections. Medical mycology 2012; 50(8): 785-94.11. Lanternier F, Cypowyj S, Picard C, et al. Primary immunodeficiencies underlying fungal infections. Curr Opin Pediatr 2013; 25(6): 736-47.12. van de Veerdonk FL, Netea MG, Joosten LA, van der Meer JW, Kullberg BJ. Novel strategies for the prevention and treatment of Candida infections: the potential of immunotherapy. FEMS Microbiol Rev 2010; 34(6): 1063-75.13. Vazquez JA, Gupta S, Villanueva A. Potential utility of recombinant human GM-CSF as adjunctive treatment of refractory oropharyngeal candidiasis in AIDS patients. Eur J Clin Microbiol Infect Dis 1998; 17(11): 781-3.14. Vazquez JA, Hidalgo JA, De Bono S. Use of sargramostim (rh-GM-CSF) as adjunctive treatment of fluconazole-refractory oropharyngeal candidiasis in patients with AIDS: a pilot study. HIV Clin Trials 2000; 1(3): 23-9.15. Wan L, Zhang Y, Lai Y, et al. Effect of Granulocyte-Macrophage Colony-Stimulating Factor on Prevention and Treatment of Invasive Fungal Disease in Recipients of Allogeneic Stem-Cell Transplantation: A Prospective Multicenter Randomized Phase IV Trial. J Clin Oncol 2015; 33(34): 3999-4006.16. Bandera A, Trabattoni D, Ferrario G, et al. Interferon-gamma and granulocyte-macrophage colony stimulating factor therapy in three patients with pulmonary aspergillosis. Infection 2008; 36(4): 368-73.17. Saijo T, Chen J, Chen SC, et al. Anti-granulocyte-macrophage colony-stimulating factor autoantibodies are a risk factor for central nervous system infection by Cryptococcus gattii in otherwise immunocompetent patients. mBio 2014; 5(2): e00912-14.18. Kullberg BJ, Oude Lashof AM, Netea MG. Design of efficacy trials of cytokines in combination with antifungal drugs. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 2004; 39 Suppl 4: S218-23.

Page 27: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

19. Todeschini G, Murari C, Bonesi R, et al. Invasive aspergillosis in neutropenic patients: rapid neutrophil recovery is a risk factor for severe pulmonary complications. Eur J Clin Invest 1999; 29(5): 453-7.20. Kullberg BJ, van 't Wout JW, Hoogstraten C, van Furth R. Recombinant interferon-gamma enhances resistance to acute disseminated Candida albicans infection in mice. Journal of Infectious Diseases 1993; 168(2): 436-43.21. Nagai H, Guo J, Choi H, Kurup V. Interferon-gamma and tumor necrosis factor-alpha protect mice from invasive aspergillosis. J Infect Dis 1995; 172(6): 1554-60.22. Clemons KV, Lutz JE, Stevens DA. Efficacy of recombinant gamma interferon for treatment of systemic cryptococcosis in SCID mice. Antimicrob Agents Chemother 2001; 45(3): 686-9.23. A Controlled Trial of Interferon Gamma to Prevent Infection in Chronic Granulomatous Disease. New England Journal of Medicine 1991; 324(8): 509-16.24. Marciano BE, Wesley R, De Carlo ES, et al. Long-term interferon-gamma therapy for patients with chronic granulomatous disease. Clinical infectious diseases 2004; 39(5): 692-9.25. Armstrong-James D, Teo IA, Shrivastava S, et al. Exogenous interferon-gamma immunotherapy for invasive fungal infections in kidney transplant patients. Am J Transplant 2010; 10(8): 1796-803.26. Siddiqui AA, Brouwer AE, Wuthiekanun V, et al. IFN-gamma at the site of infection determines rate of clearance of infection in cryptococcal meningitis. J Immunol 2005; 174(3): 1746-50.27. Jarvis JN, Meintjes G, Rebe K, et al. Adjunctive interferon-gamma immunotherapy for the treatment of HIV-associated cryptococcal meningitis: a randomized controlled trial. Aids 2012; 26(9): 1105-13.28. Netea MG, Brouwer AE, Hoogendoorn EH, et al. Two patients with cryptococcal meningitis and idiopathic CD4 lymphopenia: defective cytokine production and reversal by recombinant interferon- gamma therapy. Clinical infectious diseases 2004; 39(9): e83-7.29. Delsing CE, Gresnigt MS, Leentjens J, et al. Interferon-gamma as adjunctive immunotherapy for invasive fungal infections: a case series. BMC Infect Dis 2014; 14: 166.30. Meintjes G, Scriven J, Marais S. Management of the immune reconstitution inflammatory syndrome. Curr HIV/AIDS Rep 2012; 9(3): 238-50.31. Raberg L, Sim D, Read AF. Disentangling genetic variation for resistance and tolerance to infectious diseases in animals. Science 2007; 318(5851): 812-4.32. Zelante T, Iannitti R, De Luca A, Romani L. IL-22 in antifungal immunity. Eur J Immunol 2011; 41(2): 270-5.33. Casadevall A, Pirofski LA. The damage-response framework of microbial pathogenesis. Nat Rev Microbiol 2003; 1(1): 17-24.34. Montagnoli C, Bozza S, Gaziano R, et al. Immunity and tolerance to Aspergillus fumigatus. Novartis Found Symp 2006; 279: 66-77; discussion -9, 216-9.35. Kisand K, Lilic D, Casanova JL, Peterson P, Meager A, Willcox N. Mucocutaneous candidiasis and autoimmunity against cytokines in APECED and thymoma patients: clinical and pathogenetic implications. Eur J Immunol 2011; 41(6): 1517-27.36. Kisand K, Boe Wolff AS, Podkrajsek KT, et al. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. The Journal of experimental medicine 2010; 207(2): 299-308.37. Browne SK. Anticytokine autoantibody-associated immunodeficiency. Annu Rev Immunol 2014; 32: 635-57.38. Panackal AA, Wuest SC, Lin YC, et al. Paradoxical Immune Responses in Non-HIV Cryptococcal Meningitis. PLoS Pathog 2015; 11(5): e1004884.39. Tang BS, Chan JF, Chen M, et al. Disseminated penicilliosis, recurrent bacteremic nontyphoidal salmonellosis, and burkholderiosis associated with acquired immunodeficiency due to autoantibody against gamma interferon. Clinical and vaccine immunology : CVI 2010; 17(7): 1132-8.

Page 28: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

40. Browne SK, Zaman R, Sampaio EP, et al. Anti-CD20 (rituximab) therapy for anti-IFN-gamma autoantibody-associated nontuberculous mycobacterial infection. Blood 2012; 119(17): 3933-9.41. Legrand F, Lecuit M, Dupont B, et al. Adjuvant corticosteroid therapy for chronic disseminated candidiasis. Clin Infect Dis 2008; 46(5): 696-702.42. Ferwerda B, Ferwerda G, Plantinga TS, et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N Engl J Med 2009; 361(18): 1760-7.43. Borghi M, De Luca A, Puccetti M, et al. Pathogenic NLRP3 Inflammasome Activity during Candida Infection Is Negatively Regulated by IL-22 via Activation of NLRC4 and IL-1Ra. Cell Host Microbe 2015; 18(2): 198-209.44. Majer O, Bourgeois C, Zwolanek F, et al. Type I interferons promote fatal immunopathology by regulating inflammatory monocytes and neutrophils during Candida infections. PLoS Pathog 2012; 8(7): e1002811.45. Ling Y, Cypowyj S, Aytekin C, et al. Inherited IL-17RC deficiency in patients with chronic mucocutaneous candidiasis. J Exp Med 2015; 212(5): 619-31.46. Boisson B, Wang C, Pedergnana V, et al. An ACT1 mutation selectively abolishes interleukin-17 responses in humans with chronic mucocutaneous candidiasis. Immunity 2013; 39(4): 676-86.47. Milner JD, Sandler NG, Douek DC. Th17 cells, Job's syndrome and HIV: opportunities for bacterial and fungal infections. Curr Opin HIV AIDS 2010; 5(2): 179-83.48. Toubiana J, Okada S, Hiller J, et al. Heterozygous STAT1 gain-of-function mutations underlie an unexpectedly broad clinical phenotype. Blood 2016; 127(25): 3154-64.49. Mossner R, Diering N, Bader O, et al. Ruxolitinib Induces Interleukin 17 and Ameliorates Chronic Mucocutaneous Candidiasis Caused by STAT1 Gain-of-Function Mutation. Clin Infect Dis 2016; 62(7): 951-3.50. Higgins E, Al Shehri T, McAleer MA, et al. Use of ruxolitinib to successfully treat chronic mucocutaneous candidiasis caused by gain-of-function signal transducer and activator of transcription 1 (STAT1) mutation. J Allergy Clin Immunol 2015; 135(2): 551-3.51. Kyrmizi I, Gresnigt MS, Akoumianaki T, et al. Corticosteroids block autophagy protein recruitment in Aspergillus fumigatus phagosomes via targeting dectin-1/Syk kinase signaling. J Immunol 2013; 191(3): 1287-99.52. de Luca A, Smeekens SP, Casagrande A, et al. IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proceedings of the National Academy of Sciences of the United States of America 2014; 111(9): 3526-31.53. Horvath R, Rozkova D, Lastovicka J, et al. Expansion of T helper type 17 lymphocytes in patients with chronic granulomatous disease. Clin Exp Immunol 2011; 166(1): 26-33.54. Romani L, Fallarino F, De Luca A, et al. Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature 2008; 451(7175): 211-5.55. Knutsen AP, Bush RK, Demain JG, et al. Fungi and allergic lower respiratory tract diseases. J Allergy Clin Immunol 2012; 129(2): 280-91; quiz 92-3.56. Chishimba L, Niven RM, Cooley J, Denning DW. Voriconazole and posaconazole improve asthma severity in allergic bronchopulmonary aspergillosis and severe asthma with fungal sensitization. The Journal of asthma : official journal of the Association for the Care of Asthma 2012; 49(4): 423-33.57. Latzin P, Hartl D, Regamey N, Frey U, Schoeni MH, Casaulta C. Comparison of serum markers for allergic bronchopulmonary aspergillosis in cystic fibrosis. The European respiratory journal 2008; 31(1): 36-42.58. Hartl D, Buckland KF, Hogaboam CM. Chemokines in allergic aspergillosis--from animal models to human lung diseases. Inflammation & allergy drug targets 2006; 5(4): 219-28.59. Dubey LK, Moeller JB, Schlosser A, Sorensen GL, Holmskov U. Induction of innate immunity by Aspergillus fumigatus cell wall polysaccharides is enhanced by the composite presentation of chitin and beta-glucan. Immunobiology 2014; 219(3): 179-88.

Page 29: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

60. Hartl D. Immunological mechanisms behind the cystic fibrosis-ABPA link. Medical mycology : official publication of the International Society for Human and Animal Mycology 2009; 47 Suppl 1: S183-91.61. Nguyen NL, Chen K, McAleer J, Kolls JK. Vitamin D regulation of OX40 ligand in immune responses to Aspergillus fumigatus. Infection and immunity 2013; 81(5): 1510-9.62. Nguyen NL, Pilewski JM, Celedon JC, et al. Vitamin D supplementation decreases specific Th2 responses in CF patients with aspergillus sensitization: a phase one open-label study. Asthma Res Pract 2015; 1.63. Wong R, Wong M, Robinson PD, Fitzgerald DA. Omalizumab in the management of steroid dependent allergic bronchopulmonary aspergillosis (ABPA) complicating cystic fibrosis. Paediatric respiratory reviews 2013; 14(1): 22-4.64. Homma T, Kurokawa M, Matsukura S, Yamaguchi M, Adachi M. Anti-IgE therapy for allergic bronchopulmonary aspergillosis. Journal of microbiology, immunology, and infection = Wei mian yu gan ran za zhi 2013.65. Collins J, Devos G, Hudes G, Rosenstreich D. Allergic bronchopulmonary aspergillosis treated successfully for one year with omalizumab. Journal of asthma and allergy 2012; 5: 65-70.66. Aydin O, Sozener ZC, Soyyigit S, et al. Omalizumab in the treatment of allergic bronchopulmonary aspergillosis: One center's experience with 14 cases. Allergy and asthma proceedings : the official journal of regional and state allergy societies 2015; 36(6): 493-500.67. Becker KL, Gresnigt MS, Smeekens SP, et al. Pattern recognition pathways leading to a Th2 cytokine bias in ABPA patients. Clin Exp Allergy 2014.68. Castanhinha S, Sherburn R, Walker S, et al. Pediatric severe asthma with fungal sensitization is mediated by steroid-resistant IL-33. Journal of Allergy and Clinical Immunology 2015; 136(2): 312-22.e7.69. Fei M, Bhatia S, Oriss TB, et al. TNF-alpha from inflammatory dendritic cells (DCs) regulates lung IL-17A/IL-5 levels and neutrophilia versus eosinophilia during persistent fungal infection. Proceedings of the National Academy of Sciences of the United States of America 2011; 108(13): 5360-5.70. Levitz SM, Golenbock DT. Beyond empiricism: informing vaccine development through innate immunity research. Cell 2012; 148(6): 1284-92.71. Torosantucci A, Bromuro C, Chiani P, et al. A novel glyco-conjugate vaccine against fungal pathogens. J Exp Med 2005; 202(5): 597-606.72. Specht CA, Lee CK, Huang H, et al. Protection against Experimental Cryptococcosis following Vaccination with Glucan Particles Containing Cryptococcus Alkaline Extracts. mBio 2015; 6(6): e01905-15.73. Santos E, Levitz SM. Fungal vaccines and immunotherapeutics. Cold Spring Harb Perspect Med 2014; 4(11): a019711.74. Wozniak KL, Young ML, Wormley FL, Jr. Protective immunity against experimental pulmonary cryptococcosis in T cell-depleted mice. Clinical and vaccine immunology : CVI 2011; 18(5): 717-23.75. Pappagianis D, Group TVFVS. Evaluation of the protective efficacy of the killed Coccidioides immitis spherule vaccine in humans. American Review of Respiratory Disease 1993; 148: 656-60.76. Schmidt CS, White CJ, Ibrahim AS, et al. NDV-3, a recombinant alum-adjuvanted vaccine for Candida and Staphylococcus aureus, is safe and immunogenic in healthy adults. Vaccine 2012; 30(52): 7594-600.77. De Bernardis F, Amacker M, Arancia S, et al. A virosomal vaccine against candidal vaginitis: immunogenicity, efficacy and safety profile in animal models. Vaccine 2012; 30(30): 4490-8.78. McClelland EE, Nicola AM, Prados-Rosales R, Casadevall A. Ab binding alters gene expression in Cryptococcus neoformans and directly modulates fungal metabolism. J Clin Invest 2010; 120(4): 1355-61.

Page 30: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

79. Larsen RA, Pappas PG, Perfect J, et al. Phase I evaluation of the safety and pharmacokinetics of murine-derived anticryptococcal antibody 18B7 in subjects with treated cryptococcal meningitis. Antimicrob Agents Chemother 2005; 49(3): 952-8.80. Delorme EJ, Alexander P. Treatment of Primary Fibrosarcoma in the Rat with Immune Lymphocytes. Lancet 1964; 2(7351): 117-20.81. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A 1989; 86(24): 10024-8.82. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015; 348(6230): 62-8.83. Dale DC, Liles WC, Price TH. Renewed interest in granulocyte transfusion therapy. Br J Haematol 1997; 98(3): 497-501.84. Beck O, Koehl U, Tramsen L, et al. Enumeration of functionally active anti-Aspergillus T-cells in human peripheral blood. J Immunol Methods 2008; 335(1-2): 41-5.85. Perruccio K, Tosti A, Burchielli E, et al. Transferring functional immune responses to pathogens after haploidentical hematopoietic transplantation. Blood 2005; 106(13): 4397-406.86. Papadopoulou A, Kaloyannidis P, Yannaki E, Cruz CR. Adoptive transfer of Aspergillus-specific T cells as a novel anti-fungal therapy for hematopoietic stem cell transplant recipients: Progress and challenges. Crit Rev Oncol Hematol 2016; 98: 62-72.87. Tramsen L, Schmidt S, Boenig H, et al. Clinical-scale generation of multi-specific anti-fungal T cells targeting Candida, Aspergillus and mucormycetes. Cytotherapy 2013; 15(3): 344-51.88. Srivastava S, Riddell SR. Engineering CAR-T cells: Design concepts. Trends Immunol 2015; 36(8): 494-502.89. Kumaresan PR, Manuri PR, Albert ND, et al. Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection. Proc Natl Acad Sci U S A 2014; 111(29): 10660-5.90. Seidel MG, Peters C, Wacker A, et al. Randomized phase III study of granulocyte transfusions in neutropenic patients. Bone Marrow Transplant 2008; 42(10): 679-84.91. Price TH, Boeckh M, Harrison RW, et al. Efficacy of transfusion with granulocytes from G-CSF/dexamethasone-treated donors in neutropenic patients with infection. Blood 2015; 126(18): 2153-61.92. Bozza S, Perruccio K, Montagnoli C, et al. A dendritic cell vaccine against invasive aspergillosis in allogeneic hematopoietic transplantation. Blood 2003; 102(10): 3807-14.93. Schmidt S, Tramsen L, Hanisch M, et al. Human natural killer cells exhibit direct activity against Aspergillus fumigatus hyphae, but not against resting conidia. J Infect Dis 2011; 203(3): 430-5.94. Schmidt S, Tramsen L, Perkhofer S, et al. Rhizopus oryzae hyphae are damaged by human natural killer (NK) cells, but suppress NK cell mediated immunity. Immunobiology 2013; 218(7): 939-44.95. Stern M, Passweg JR, Meyer-Monard S, et al. Pre-emptive immunotherapy with purified natural killer cells after haploidentical SCT: a prospective phase II study in two centers. Bone Marrow Transplant 2013; 48(3): 433-8.96. Sousa Mda G, Reid DM, Schweighoffer E, et al. Restoration of pattern recognition receptor costimulation to treat chromoblastomycosis, a chronic fungal infection of the skin. Cell Host Microbe 2011; 9(5): 436-43.97. de Sousa Mda G, Belda W, Jr., Spina R, et al. Topical application of imiquimod as a treatment for chromoblastomycosis. Clinical infectious diseases 2014; 58(12): 1734-7.98. Maskarinec SA, Johnson MD, Perfect JR. Genetic Susceptibility to Fungal Infections: What is in the Genes? Curr Clin Microbiol Rep 2016; 3(2): 81-91.99. Wojtowicz A, Bochud PY. Host genetics of invasive Aspergillus and Candida infections. Semin Immunopathol 2015; 37(2): 173-86.

Page 31: Imperial College London · Web viewCellular immunotherapy Cellular therapy represents a promising approach to treating infection, by redirecting the power of immune cells through

100. Armstrong-James D, Teo I, Herbst S, et al. Renal allograft recipients fail to increase interferon-gamma during invasive fungal diseases. Am J Transplant 2012; 12(12): 3437-40.101. Netea MG, Brouwer AE, Hoogendoorn EH, et al. Two patients with cryptococcal meningitis and idiopathic CD4 lymphopenia: defective cytokine production and reversal by recombinant interferon- gamma therapy. Clin Infect Dis 2004; 39(9): e83-7.