Advertisement
Original Article|Articles in Press

Endolysin inhibits skin colonization by patient-derived Staphylococcus aureus and malignant T cell activation in cutaneous T cell lymphoma

Open AccessPublished:March 06, 2023DOI:https://doi.org/10.1016/j.jid.2023.01.039

      ABSTRACT

      Staphylococcus aureus (S. aureus) is suspected to fuel disease activity in cutaneous T cell lymphomas (CTCL). Here we investigate the effect of a recombinant, anti-bacterial protein, endolysin, XZ.700, on S. aureus skin colonization and malignant T cell activation. We show that endolysin strongly inhibits proliferation of S. aureus isolated from CTCL skin and significantly decreases S. aureus bacterial cell counts in a dose-dependent manner. Likewise, ex vivo colonization of both healthy and lesional skin by S. aureus is profoundly inhibited by endolysin. Moreover, endolysin inhibits the patient-derived S. aureus induction of Interferon-gamma (IFNγ) and IFNγ-inducible chemokine CXCL10 in healthy skin. Whereas patient-derived S. aureus stimulates activation and proliferation of malignant T cells in vitro through an indirect mechanism involving non-malignant T cells, endolysin strongly inhibits the effects of S. aureus on activation (reduced CD25 and STAT5 phosphorylation) and proliferation (reduced Ki67) of malignant T cells and cell lines in the presence of non-malignant T cells. Taken together, we provide evidence that endolysin XZ.700 inhibits skin colonization, chemokine expression, and proliferation of pathogenic S. aureus, and blocks their potential tumor-promoting effects on malignant T cells.

      INTRODUCTION

      Cutaneous T-cell lymphomas (CTCL) are a heterogeneous group of non-Hodgkin lymphomas, with mycosis fungoides (MF) and Sézary syndrome (SS) being the most prevalent and most severe subtypes, respectively (
      • Aschebrook-Kilfoy B
      • Cocco P
      • La Vecchia C
      • Chang ET
      • Vajdic CM
      • Kadin ME
      • et al.
      Medical history, lifestyle, family history, and occupational risk factors for mycosis fungoides and Sezary syndrome: the InterLymph Non-Hodgkin Lymphoma Subtypes Project.
      ,
      • Dobos G
      • de Masson A
      • Ram-Wolff C
      • Beylot-Barry M
      • Pham-Ledard A
      • Ortonne N
      • et al.
      Epidemiological changes in cutaneous lymphomas: an analysis of 8593 patients from the French Cutaneous Lymphoma Registry.
      ,
      • Hodak E
      • Amitay-Laish I
      Mycosis fungoides: A great imitator.
      ). Multiple cancer cell-intrinsic genetic events and epigenetic regulators seem to play an important role in CTCL pathogenesis, however, a complete understanding of disease etiology and pathogenesis remains elusive (
      • Dummer R
      • Vermeer MH
      • Scarisbrick JJ
      • Kim YH
      • Stonesifer C
      • Tensen CP
      • et al.
      Cutaneous T cell lymphoma.
      ,
      • Gluud M
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Lindahl LM
      • Buus TB
      • Andersen MH
      • et al.
      MicroRNAs in the Pathogenesis, Diagnosis, Prognosis and Targeted Treatment of Cutaneous T-Cell Lymphomas.
      ,
      • Qu K
      • Zaba LC
      • Satpathy AT
      • Giresi PG
      • Li R
      • Jin Y
      • et al.
      Chromatin Accessibility Landscape of Cutaneous T Cell Lymphoma and Dynamic Response to HDAC Inhibitors.
      ,
      • Tensen CP
      • Quint KD
      • Vermeer MH
      Genetic and epigenetic insights into cutaneous T-cell lymphoma.
      ,
      • van Doorn R
      • Slieker RC
      • Boonk SE
      • Zoutman WH
      • Goeman JJ
      • Bagot M
      • et al.
      Epigenomic Analysis of Sezary Syndrome Defines Patterns of Aberrant DNA Methylation and Identifies Diagnostic Markers.
      ). Recent focus has turned to the interplay between malignant T cells, benign T cells, and stromal cells, and how these interactions shape the tumor microenvironment (TME) in ways that promote disease progression and weaken the anti-tumor and anti-microbial immune defense (
      • Bagot M
      • Nikolova M
      • Schirm-Chabanette F
      • Wechsler J
      • Boumsell L
      • Bensussan A
      Crosstalk between tumor T lymphocytes and reactive T lymphocytes in cutaneous T cell lymphomas.
      ,
      • Dobos G
      • Calugareanu A
      • Michel L
      • Battistella M
      • Ram-Wolff C
      • Bouaziz JD
      • et al.
      Exploring the role of the skin microenvironment in cutaneous T-cell lymphoma using single cell RNA-sequencing.
      ,
      • Edelson RL
      Cutaneous T cell lymphoma: the helping hand of dendritic cells.
      ,
      • Herrera A
      • Cheng A
      • Mimitou EP
      • Seffens A
      • George D
      • Bar-Natan M
      • et al.
      Multimodal single-cell analysis of cutaneous T-cell lymphoma reveals distinct subclonal tissue-dependent signatures.
      ,
      • Krejsgaard T
      • Lindahl LM
      • Mongan NP
      • Wasik MA
      • Litvinov IV
      • Iversen L
      • et al.
      Malignant inflammation in cutaneous T-cell lymphoma-a hostile takeover.
      ,
      • Mehdi SJ
      • Moerman-Herzog A
      • Wong HK
      Normal and cancer fibroblasts differentially regulate TWIST1, TOX and cytokine gene expression in cutaneous T-cell lymphoma.
      ,
      • Vieyra-Garcia P
      • Crouch JD
      • O'Malley JT
      • Seger EW
      • Yang CH
      • Teague JE
      • et al.
      Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma.
      ,
      • Wu X
      • Singh R
      • Hsu DK
      • Zhou Y
      • Yu S
      • Han D
      • et al.
      A Small Molecule CCR2 Antagonist Depletes Tumor Macrophages and Synergizes with Anti-PD-1 in a Murine Model of Cutaneous T-Cell Lymphoma (CTCL).
      ,
      • Yu KK
      • Smith NP
      • Essien SV
      • Teague JE
      • Vieyra-Garcia P
      • Gehad A
      • et al.
      IL-32 supports the survival of malignant T cells in cutaneous T cell lymphoma.
      ,
      • Gluud M
      • Pallesen EMH
      • Buus TB
      • Gjerdrum LMR
      • Lindahl LM
      • Kamstrup MR
      • et al.
      Malignant T cells induce skin barrier defects through cytokine-mediated JAK/STAT signalling in cutaneous T-cell lymphoma.
      ). The cellular interactions are among other affected by soluble factors in the TME, such as cytokines and chemokines. Chemokines are believed to have an important pathogenetic role in CTCL, as chemokines and their receptors are crucial for homing of malignant T cells and immune cells to the skin (
      • de Masson A
      • Darbord D
      • Dobos G
      • Boisson M
      • Roelens M
      • Ram-Wolff C
      • et al.
      Macrophage-derived CXCL9 and CXCL11, T-cell skin homing, and disease control in mogamulizumab-treated CTCL patients.
      ,
      • Geskin LJ
      • Akilov OE
      • Lin Y
      • Lokshin AE
      Distinct age-matched serum biomarker profiles in patients with cutaneous T-cell lymphoma.
      ,
      • Stolearenco V
      • Namini MRJ
      • Hasselager SS
      • Gluud M
      • Buus TB
      • Willerslev-Olsen A
      • et al.
      Cellular Interactions and Inflammation in the Pathogenesis of Cutaneous T-Cell Lymphoma.
      ,
      • Wu XS
      • Lonsdorf AS
      • Hwang ST
      Cutaneous T-cell lymphoma: roles for chemokines and chemokine receptors.
      ). Pionering studies in the 1990’s indicated that the Interferon-gamma (IFNγ)-inducible chemokine CXCL10 (IP10) plays a key role in homing of CD4 T cells to the epidermis (
      • Sarris AH
      • Daliani D
      • Ulmer R
      • Crow M
      • Broxmeyer HE
      • Pugh W
      • et al.
      Interferon-inducible protein-10 and the pathogenesis of cutaneous T-cell lymphomas.
      ,
      • Sarris AH
      • Esgleyes-Ribot T
      • Crow M
      • Broxmeyer HE
      • Karasavvas N
      • Pugh W
      • et al.
      Cytokine loops involving interferon-gamma and IP-10, a cytokine chemotactic for CD4+ lymphocytes: an explanation for the epidermotropism of cutaneous T-cell lymphoma?.
      ,
      • Tensen CP
      • Vermeer MH
      • van der Stoop PM
      • van Beek P
      • Scheper RJ
      • Boorsma DM
      • et al.
      Epidermal interferon-gamma inducible protein-10 (IP-10) and monokine induced by gamma-interferon (Mig) but not IL-8 mRNA expression is associated with epidermotropism in cutaneous T cell lymphomas.
      ), a phenomenon named epidermotropism, which is a characteristic feature of CTCL (
      • Edelson RL
      Cutaneous T cell lymphoma: mycosis fungoides, Sezary syndrome, and other variants.
      ). Later investigations have revealed that numerous other chemokines and chemokine-receptors are also involved in the pathogenesis of CTCL (reviewed in (
      • de Masson A
      • Darbord D
      • Dobos G
      • Boisson M
      • Roelens M
      • Ram-Wolff C
      • et al.
      Macrophage-derived CXCL9 and CXCL11, T-cell skin homing, and disease control in mogamulizumab-treated CTCL patients.
      ,
      • Stolearenco V
      • Namini MRJ
      • Hasselager SS
      • Gluud M
      • Buus TB
      • Willerslev-Olsen A
      • et al.
      Cellular Interactions and Inflammation in the Pathogenesis of Cutaneous T-Cell Lymphoma.
      ,
      • Wu XS
      • Lonsdorf AS
      • Hwang ST
      Cutaneous T-cell lymphoma: roles for chemokines and chemokine receptors.
      ). Notably, CCR4 antibodies, such as the CCR4 blocker mogamulizumab, have shown treatment efficacy in CTCL (approved for relapsed or refractory MF/SS) (
      • Kim YH
      • Bagot M
      • Pinter-Brown L
      • Rook AH
      • Porcu P
      • Horwitz SM
      • et al.
      Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): an international, open-label, randomised, controlled phase 3 trial.
      ).
      An important feature of CTCL is an increased susceptibility to bacterial infections, which are common in CTCL (
      • Axelrod PI
      • Lorber B
      • Vonderheid EC
      Infections complicating mycosis fungoides and Sezary syndrome.
      ,
      • Jackow CM
      • Cather JC
      • Hearne V
      • Asano AT
      • Musser JM
      • Duvic M
      Association of erythrodermic cutaneous T-cell lymphoma, superantigen-positive Staphylococcus aureus, and oligoclonal T-cell receptor V beta gene expansion.
      ,
      • Kadin ME
      • Hamilton RG
      • Vonderheid EC
      Evidence linking atopy and staphylococcal superantigens to the pathogenesis of lymphomatoid papulosis, a recurrent CD30+ cutaneous lymphoproliferative disorder.
      ) and a major cause of disease morbidity and mortality (
      • Axelrod PI
      • Lorber B
      • Vonderheid EC
      Infections complicating mycosis fungoides and Sezary syndrome.
      ,
      • Emge DA
      • Bassett RL
      • Duvic M
      • Huen AO
      Methicillin-resistant Staphylococcus aureus (MRSA) is an important pathogen in erythrodermic cutaneous T-cell lymphoma (CTCL) patients.
      ,
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ,
      • Mirvish ED
      • Pomerantz RG
      • Geskin LJ
      Infectious agents in cutaneous T-cell lymphoma.
      ). Skin barrier defects induced by the malignant T cells are likely ports of entrance for bacteria (
      • Gluud M
      • Pallesen EMH
      • Buus TB
      • Gjerdrum LMR
      • Lindahl LM
      • Kamstrup MR
      • et al.
      Malignant T cells induce skin barrier defects through cytokine-mediated JAK/STAT signalling in cutaneous T-cell lymphoma.
      ). Even without overt bacterial infection, the skin microbiota may play a role in CTCL severity. This is exemplified by the reduced disease severity and tumor burden following systemic antibiotics therapy used either prophylactic or in relation to non-cutaneous infections (
      • Lewis DJ
      • Holder BB
      • Duvic M
      The "Duvic regimen" for erythrodermic flares secondary to Staphylococcus aureus in mycosis fungoides and Sezary syndrome.
      ,
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ,
      • Talpur R
      • Bassett R
      • Duvic M
      Prevalence and treatment of Staphylococcus aureus colonization in patients with mycosis fungoides and Sezary syndrome.
      ,
      • Tokura Y
      • Yagi H
      • Ohshima A
      • Kurokawa S
      • Wakita H
      • Yokote R
      • et al.
      Cutaneous colonization with staphylococci influences the disease activity of Sezary syndrome: a potential role for bacterial superantigens.
      ). Here S. aureus and staphylococcal enterotoxins (SE) have been suspected to fuel disease activity and cancer progression in CTCL through a direct stimulation of the malignant T cell clone and/or through modulation of the TME (
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ,
      • Tokura Y
      • Yagi H
      • Ohshima A
      • Kurokawa S
      • Wakita H
      • Yokote R
      • et al.
      Cutaneous colonization with staphylococci influences the disease activity of Sezary syndrome: a potential role for bacterial superantigens.
      ,
      • Willerslev-Olsen A
      • Krejsgaard T
      • Lindahl LM
      • Bonefeld CM
      • Wasik MA
      • Koralov SB
      • et al.
      Bacterial toxins fuel disease progression in cutaneous T-cell lymphoma.
      ). These findings are supported by gnotobiotic experiments in a murine CTCL disease model, showing that fulminant disease development did not develop in germ free settings (
      • Fanok MH
      • Sun A
      • Fogli LK
      • Narendran V
      • Eckstein M
      • Kannan K
      • et al.
      Role of Dysregulated Cytokine Signaling and Bacterial Triggers in the Pathogenesis of Cutaneous T-Cell Lymphoma.
      ). Importantly, S. aureus toxins trigger signaling pathways associated with CTCL pathogenesis in peripheral blood mononuclear cells (PBMC) isolated from Sézary syndrome patients with both malignant and non-malignant T cells: (i) expression of oncogenic microRNA-155 and regulatory proteins (PD1, FOXP3, and IL-10), (ii) STAT3 and STAT5 activation, (iii) inhibition of anti-tumor cytotoxicity, and (iv) proliferation of primary malignant T cells (
      • Blumel E
      • Munir Ahmad S
      • Nastasi C
      • Willerslev-Olsen A
      • Gluud M
      • Fredholm S
      • et al.
      Staphylococcus aureus alpha-toxin inhibits CD8(+) T cell-mediated killing of cancer cells in cutaneous T-cell lymphoma.
      ,
      • Blumel E
      • Willerslev-Olsen A
      • Gluud M
      • Lindahl LM
      • Fredholm S
      • Nastasi C
      • et al.
      Staphylococcal alpha-toxin tilts the balance between malignant and non-malignant CD4(+) T cells in cutaneous T-cell lymphoma.
      ,
      • Krejsgaard T
      • Litvinov IV
      • Wang Y
      • Xia L
      • Willerslev-Olsen A
      • Koralov SB
      • et al.
      Elucidating the role of interleukin-17F in cutaneous T-cell lymphoma.
      ,
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ,
      • Willerslev-Olsen A
      • Buus TB
      • Nastasi C
      • Blumel E
      • Gluud M
      • Bonefeld CM
      • et al.
      Staphylococcus aureus enterotoxins induce FOXP3 in neoplastic T cells in Sezary syndrome.
      ,
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Lindahl LM
      • Buus TB
      • Pallesen EMH
      • Gluud M
      • et al.
      Staphylococcus aureus Induces Signal Transducer and Activator of Transcription 5Dependent miR-155 Expression in Cutaneous T-Cell Lymphoma.
      ,
      • Willerslev-Olsen A
      • Krejsgaard T
      • Lindahl LM
      • Litvinov IV
      • Fredholm S
      • Petersen DL
      • et al.
      Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma.
      ). Together, this supports the hypothesis of a mechanistic link between S. aureus and cancer progression in CTCL (
      • Jackow CM
      • Cather JC
      • Hearne V
      • Asano AT
      • Musser JM
      • Duvic M
      Association of erythrodermic cutaneous T-cell lymphoma, superantigen-positive Staphylococcus aureus, and oligoclonal T-cell receptor V beta gene expansion.
      ,
      • Tokura Y
      • Heald PW
      • Yan SL
      • Edelson RL
      Stimulation of cutaneous T-cell lymphoma cells with superantigenic staphylococcal toxins.
      ,
      • Tokura Y
      • Yagi H
      • Ohshima A
      • Kurokawa S
      • Wakita H
      • Yokote R
      • et al.
      Cutaneous colonization with staphylococci influences the disease activity of Sezary syndrome: a potential role for bacterial superantigens.
      ,
      • Willerslev-Olsen A
      • Krejsgaard T
      • Lindahl LM
      • Bonefeld CM
      • Wasik MA
      • Koralov SB
      • et al.
      Bacterial toxins fuel disease progression in cutaneous T-cell lymphoma.
      ). Nevertheless, re-colonization of skin lesions by S. aureus is seen in most CTCL patients after termination of antibiotic treatment indicative of the need for a lasting regulation of disease promoting bacteria, such as S. aureus on the skin (
      • Lindahl LM
      • Iversen L
      • Odum N
      • Kilian M
      Staphylococcus aureus and Antibiotics in Cutaneous T-Cell Lymphoma.
      ). Long-term usage of antibiotics to persue this goal comes with the risk of developing drug resistance which is not warranted in a patient group already susceptible to infections thereby making it even more difficult to treat infections requiring medical attention. Thus, there is a medical need for new, specific anti-staphylococcal alternatives to the currently used antibiotics.
      Recombinantly produced endolysins represent an interesting new class of antibacterial agents. Endolysins are peptidoglycan (PG) hydrolases expressed by bacteriophages. As endolysins cleave the bonds within the PG, they can target both metabolically active and inactive bacteria, such as bacterial biofilms (
      • Kuiper JWP
      • Hogervorst JMA
      • Herpers BL
      • Bakker AD
      • Klein-Nulend J
      • Nolte PA
      • et al.
      The novel endolysin XZ.700 effectively treats MRSA biofilms in two biofilm models without showing toxicity on human bone cells in vitro.
      ,
      • Schmelcher M
      • Shen Y
      • Nelson DC
      • Eugster MR
      • Eichenseher F
      • Hanke DC
      • et al.
      Evolutionarily distinct bacteriophage endolysins featuring conserved peptidoglycan cleavage sites protect mice from MRSA infection.
      ). Bacterial species show unique PG compositions that can be specifically targeted by endolysins, and thereby enabling species-specificity of antimicrobial action, such as S. aureus including MRSA, while sparing the remaining commensal microbiota (
      • Eichenseher F
      • Herpers BL
      • Badoux P
      • Leyva-Castillo JM
      • Geha RS
      • van der Zwart M
      • et al.
      Linker-Improved Chimeric Endolysin Selectively Kills Staphylococcus aureus In Vitro, on Reconstituted Human Epidermis, and in a Murine Model of Skin Infection.
      ,
      • Kuiper JWP
      • Hogervorst JMA
      • Herpers BL
      • Bakker AD
      • Klein-Nulend J
      • Nolte PA
      • et al.
      The novel endolysin XZ.700 effectively treats MRSA biofilms in two biofilm models without showing toxicity on human bone cells in vitro.
      ,
      • Schmelcher M
      • Donovan DM
      • Loessner MJ
      Bacteriophage endolysins as novel antimicrobials.
      ,
      • Schmelcher M
      • Loessner MJ
      Bacteriophage endolysins - extending their application to tissues and the bloodstream.
      ). Several endolysins with specificity toward S. aureus have been recombinantly produced including endolysin XZ.700. In vitro experiments using this endolysin in sublethal dosages did not lead to the development of resistance (
      • Eichenseher F
      • Herpers BL
      • Badoux P
      • Leyva-Castillo JM
      • Geha RS
      • van der Zwart M
      • et al.
      Linker-Improved Chimeric Endolysin Selectively Kills Staphylococcus aureus In Vitro, on Reconstituted Human Epidermis, and in a Murine Model of Skin Infection.
      ). By specifically targeting S. aureus and thereby maintaining the commensal microbiota, the hope is that re-colonization by S. aureus will be hampered.
      The present study was undertaken to investigate the effect of the chimeric endolysin XZ.700 on skin colonization, chemokine expression, and possible pro-tumorigenic effects of CTCL patient-derived enterotoxin-producing S. aureus.

      RESULTS

      We first determined the bactericidal effect of the recombinant endolysin XZ.700 on a S. aureus laboratorium strain (S. aureus LAB) and S. aureus isolated from lesional skin from CTCL patients (S. aureus from patient skin (S. aureus Pt)) (Lindahl et al., 2019, Willerslev-Olsen et al., 2021). Endolysin effectively inhibited planktonic growth of S. aureus LAB (Fig. 1a) as well as a S. aureus Pt (Fig. 1b) as measured by optical density (OD). Moreover, endolysin killed both S. aureus LAB and S. aureus Pt in a dosage responsive manner with a reduction by more than four log-units at and above 1 μg/mL as determined by colony forming unit (CFU) (Fig. 1c and d). Two additional patient-derived S. aureus isolates showed similar results (Fig. 1e). As expected, the killing was abrogated by prior heat-inactivation of the endolysin (Fig. 1f) supporting that the effect is mediated by the enzymatic activity of endolysin.
      Figure thumbnail gr1
      Figure 1Endolysin (XZ.700) inhibits growth of S. aureus isolated from lesional skin of CTCL patients. S. aureus LAB strain (a and c) or CTCL patient derived S. aureus (S. aureus Pt) (b and d) were grown to early log phase and further incubated in the presence of recombinant endolysin (XZ.700) at increasing dosages. Changes in optical density (OD595nm, a and b) were determined as a function of XZ.700 treatment time and concentration (n=3; whiskers show standard deviation). Colony forming units (CFU) (c and d) were determined 1 hour after adding XZ.700 to S. aureus cultures (n=3; whiskers show standard deviation). One-way ANOVA with Dunnett's multiple comparisons test, * significant at p<0.05, *** significant at p<0.001, **** significant at p<0.0001. (e) Growth inhibition by XZ.700 at 1μg/mL was tested on two additional patient S. aureus isolates, S. aureus Pt1 and S. aureus Pt2, producing SEA (n=3; whiskers show standard deviation). (f) Growth inhibition by 1μg/mL XZ.700 with and without heat inactivation (n=3; whiskers show standard deviation).
      We setup a human ex vivo skin model to determine the effect of endolysin on S. aureus Pt colonization. Healthy skin was colonized by S. aureus Pt as indicated by arrows on the H&E and FISH stainings while no bacteria was observed in the skin without S. aureus Pt added (Fig. 2a). Importantly, endolysin profoundly inhibited colonization by S. aureus Pt (Fig. 2a). This observations was quantified by CFU measurement (Fig. 2b). When 1 μg/mL of XZ.700 was added simultaneously with S. aureus Pt, almost no skin colonization was observed (Fig. 2b). In parallel, we investigated whether endolysin could also reduce already established S. aureus Pt on skin. A significant, dose-dependent reduction of bacteria was observed for all concentrations with an almost complete eradication of S. aureus Pt using 10μg/mL (Fig. 2b). Taken together, these data provide evidence that endolysin has a potent inhibitory effect on S. aureus Pt growing on human skin.
      Figure thumbnail gr2
      Figure 2Endolysin (XZ.700) inhibits ex vivo S. aureus colonization of healthy skin and kill established S. aureus. CTCL patient-derived S. aureus were grown to early log phase and seeded on healthy skin with or without recombinant endolysin (XZ.700). (a) Micrographs depict healthy human skin without addition of S. aureus (left), addition of S. aureus Pt without XZ.700 (middle) or with co-addition of XZ.700 (right) (1μg/mL). Adhering bacteria are indicated by arrowheads. Sections are stained with H&E (upper panel) or by FISH with a S. aureus specific peptide nucleic acid (PNA) probe (lower panel). Scale bars represents 50 μm. (b) Preventing S. aureus Pt skin colonization and killing of established S. aureus Pt on human skin by XZ.700. S. aureus Pt was seeded to the stratum corneum of healthy skin specimens and incubated for 2 hours, washed with PBS to remove non-adhering S. aureus, and incubated for an additional 2 hours prior to analysis of the CFU numbers by serial dilution. XZ.700 was added together with S. aureus Pt (XZ.700CO) at 1μg/mL or after the PBS wash in concentrations 1, 5, and 10 μg/mL (XZ.700POST). Closed circles indicate number of CFU per biopsy (12.6 mm2). Open circles indicated a CFU below detection level of 100 CFU/biopsy (n=4-5; whiskers show standard deviation). Ordinary one-way ANOVA with Dunnett's multiple comparisons test, *** significant at p<0.001, **** significant at p<0.0001.
      Lesional skin from CTCL patients differs from healthy skin due to changes in skin architecture, barrier proteins, and levels of endogenous antimicrobial peptides, all of which could change S. aureus colonization and accessibility for S. aureus killing by endolysin. Accordingly, we examined the effect of endolysin on S. aureus Pt colonization of lesional skin from CTCL patients (Fig. 3). S. aureus Pt colonized the lesional CTCL skin while co-addition of endolysin significantly reduced colonization (Fig. 3). These findings suggest that the lesional skin architecture and other CTCL-associated conditions did not affect the mode of action and efficacy of endolysin on pathogenetic S. aureus.
      Figure thumbnail gr3
      Figure 3Endolysin (XZ.700) inhibits S. aureus colonization of lesional CTCL skin. Effect of co-addition of S. aureus Pt with 1 μg/mL of endolysin XZ.700 for 2.5h on colonization of lesional skin from CTCL patients. Number of living bacteria adhering to the lesional CTCL skin (CFU) from paired adjacent biopsies from a total of five lesional sites on three CTCL patients. Closed circles indicate number of CFU per biopsy (3.14 mm2). Open circles indicate CFU below the detection level of 100 CFU per biopsy. Paired t-test ** significant at p<0.01.
      As chemokines are believed to play an important pathogenetic role in CTCL, we examined the effect of S. aureus Pt on the expression of CTCL-associated chemokines in human skin. Accordingly, healthy skin specimens were incubated with S. aureus Pt supernatant for 18 hours prior to analysis of chemokine mRNA expression. As shown in figure 4a, S. aureus Pt stimulation triggered a pronounced expression of the IFNγ-inducible chemokine CXCL10, whereas the other tested CTCL-associated chemokines were not significantly upregulated (Fig. 4a). Importantly, pretreatment with endolysin partly inhibited induction of CXCL10 expression by S. aureus Pt supernatant. To validate this finding, the expression of additional S. aureus associated cytokines including IFNγ and IL-6 were analyzed. We also measured expression of IL-37, which has been inversely linked to infection-associated inflammation (
      • Allam G
      • Gaber AM
      • Othman SI
      • Abdel-Moneim A
      The potential role of interleukin-37 in infectious diseases.
      ). While IL-6 and IL-37 remained stable, incubation with S. aureus Pt supernatant induced IFNγ expression, which was strongly inhibited by endolysin (Fig. 4b). These findings suggest a novel mechanism for S. aureus to accelerate pathogenic events in CTCL, which is partially blocked by endolysin.
      Figure thumbnail gr4
      Figure 4S. aureus induced expression of CTCL-related chemokines is inhibited by endolysin (XZ.700). Relative expression (RT-qPCR) of CTCL related (a) chemokines (CXCL8, CXCL10, CCL19, CCL20, CCL22, CCL26) and (b) cytokines (IFNγ, IL-6 and IL-37) in healthy skin biopsies cultured for 18h with S. aureus Pt supernatants with and without endolysin XZ.700. mRNA expression was normalized to β-actin. The expression levels were given relative to the control (healthy skin incubated with S. aureus growth medium without addition of S. aureus (TSB)). Data are represented as an average of three independent healthy skin samples. Abbreviations: S. aureus growth medium without S. aureus (TSB); Endolysin XZ.700 diluted in TSB (XZ); S. aureus Pt supernatant (SA); S. aureus Pt supernatant with endolysin (SA+XZ).
      Toxins including SE secreted from S. aureus are suspected to change the TME (
      • Blumel E
      • Munir Ahmad S
      • Nastasi C
      • Willerslev-Olsen A
      • Gluud M
      • Fredholm S
      • et al.
      Staphylococcus aureus alpha-toxin inhibits CD8(+) T cell-mediated killing of cancer cells in cutaneous T-cell lymphoma.
      ,
      • Krejsgaard T
      • Litvinov IV
      • Wang Y
      • Xia L
      • Willerslev-Olsen A
      • Koralov SB
      • et al.
      Elucidating the role of interleukin-17F in cutaneous T-cell lymphoma.
      ,
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ,
      • Willerslev-Olsen A
      • Krejsgaard T
      • Lindahl LM
      • Litvinov IV
      • Fredholm S
      • Petersen DL
      • et al.
      Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma.
      ). Accordingly, we examined the effect of endolysin on SE production. A qualitative screening confirmed that S. aureus Pt produced staphylococcal enterotoxin A (SEA) (Suppl Fig. S1). Four hours after setting up the S. aureus Pt cultures, SEA was detected in the supernatant of S. aureus Pt at concentrations above 15 ng/mL (Fig. 5a). In contrast, the concentration of SEA in the S. aureus Pt cultures with endolysin remained below 1 ng/mL (Fig. 4a). It has previously been shown that recombinant SEA can activate non-malignant Myla1850 T cells (
      • Woetmann A
      • Lovato P
      • Eriksen KW
      • Krejsgaard T
      • Labuda T
      • Zhang Q
      • et al.
      Nonmalignant T cells stimulate growth of T-cell lymphoma cells in the presence of bacterial toxins.
      ), and thus we next addressed the effect of endolysin treated S. aureus Pt supernatants on activation of malignant and non-malignant T cells. As expected, SEA activated the non-malignant MyLa1850 T cells (CD25 expression) (Fig. 5b). Similarly, culturing Myla1850 with supernatants harvested from S. aureus Pt, resulted in T cell activation (CD25 expression) at all dilutions used (Fig. 5b). In comparison, supernatant from S. aureus Pt with 1μg/mL XZ.700 induced only weak T cell activation (Fig. 5b).
      Figure thumbnail gr5
      Figure 5Endolysin (XZ.700) inhibits S. aureus enterotoxin A production and activation of primary malignant T cells and T cell lines. (a) The concentration of SEA was determined by ELISA in supernatants from S. aureus Pt or S. aureus LAB diluted to an OD600nm of 0.01 and cultured for 4 hours with or without 1μg/mL XZ.700 (n=2). (b) In the left part, the non-malignant T cells Myla1850 were cultured in the presence of SEA (red) or unstimulated (blue) prior to flow cytometry analysis of CD25 expression. In the right part, Myla1850 were cultured in the presence of diluted supernatants from S. aureus Pt pre-treated with (blue) and without (red) endolysin at 1μg/mL prior to flow cytometry analysis of CD25 expression. (c) Co-culturing of malignant and non-malignant CTCL cells, SeAx and MyLa1850, for three days in the absence (red), presence of S. aureus enterotoxin A (blue), or supernatants harvested from S. aureus Pt treated without (orange) and with XZ.700 (green) prior to flow cytometry analysis of CD25 expression and STAT5 phosphorylation in the malignant SeAx T cells. (d) PBMCs isolated from a Sézary syndrome (SS) patient were cultured in the absence (red), presence of a pool of S. aureus enterotoxins (SEA SEB, SEC2, SED, SEI) (blue) or supernatants harvested from S. aureus Pt without (orange) and with XZ.700 (green) for three days prior to analysis by flow cytometry of CD25 expression and STAT5 phosphorylation in malignant T cells.
      Malignant SeAx T cells do not respond directly to SEA but become activated via an indirect, IL-2-dependent pathway, when exposed to SEA in co-culture with non-malignant T cells (
      • Woetmann A
      • Lovato P
      • Eriksen KW
      • Krejsgaard T
      • Labuda T
      • Zhang Q
      • et al.
      Nonmalignant T cells stimulate growth of T-cell lymphoma cells in the presence of bacterial toxins.
      ). Thus, we setup co-cultures of non-malignant Myla1850 and malignant SeAx cells and assesed T cell activation (CD25 expression and STAT5 phosphorylation). SEA and S. aureus Pt supernatant induced CD25 expression and STAT5 phosphorylation in the malignant SeAx cells (Fig. 5c), whereas pre-treatment of S. aureus Pt with endolysin inhibited supernatant-mediated T cell activation (Fig. 5c).
      We next examined the effect of S. aureus Pt supernatant and endolysin in primary malignant T cells from Sézary Syndrome patients. We used peripheral blood mononuclear cells (PBMCs), because SE-induced activation of malignant T cells usually requires crosstalk between malignant and non-malignant T cells (
      • Willerslev-Olsen A
      • Krejsgaard T
      • Lindahl LM
      • Litvinov IV
      • Fredholm S
      • Petersen DL
      • et al.
      Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma.
      ,
      • Woetmann A
      • Lovato P
      • Eriksen KW
      • Krejsgaard T
      • Labuda T
      • Zhang Q
      • et al.
      Nonmalignant T cells stimulate growth of T-cell lymphoma cells in the presence of bacterial toxins.
      ). As expected, S. aureus Pt supernatant induced CD25 expression and STAT5 phosphorylation in primary malignant T cells (Fig. 5d, brown graph). Importantly, activation of primary malignant T cells was abolished when pre-treating S. aureus Pt with endolysin (Fig. 5d, green versus brown graph).
      Next, we examined the effect of endolysin on S. aureus-mediated proliferation of malignant T cell lines from Sézary syndrome (SeAx) and mycosis fungoides (Myla3675) in co-culture with the non-malignant T cell line Myla1850. As shown in Fig. 6a, S. aureus Pt increased proliferation (Ki67 expression) of SeAx cells when cultured with MyLa1850 cells (Fig. 6a, upper row, third column) but not in SeAx cultures without MyLa1850 (Fig. 6a, upper row, first column) indicating that S. aureus Pt induced enhanced proliferation of malignant cells in co-cultures with non-malignant T cells. S. aureus Pt also increased Ki67 expression in MyLa3675 cells co-cultured with MyLa1850 cells (Fig. 6a, bottom row, third column) when compared to cultures without MyLa1850 (Fig. 6a, bottom row, first column). Of note, the high basic level of Ki67 expression in unstimulated MyLa3675 cells reflect the high level of spontaneous proliferation of this and other malignant T cell lines from this patient (
      • Krejsgaard T
      • Vetter-Kauczok CS
      • Woetmann A
      • Lovato P
      • Labuda T
      • Eriksen KW
      • et al.
      Jak3- and JNK-dependent vascular endothelial growth factor expression in cutaneous T-cell lymphoma.
      ,
      • Woetmann A
      • Lovato P
      • Eriksen KW
      • Krejsgaard T
      • Labuda T
      • Zhang Q
      • et al.
      Nonmalignant T cells stimulate growth of T-cell lymphoma cells in the presence of bacterial toxins.
      ). Importantly, endolysin almost completely blocked the effect of S. aureus Pt on Ki67 expression in both SeAx (fig. 6a, upper row, right) and MyLa3675 cells (Fig. 6a, bottom row, right) indicating that endolysin prevented S. aureus Pt enhanced proliferation of malignant T cells. This promted us to test if endolysin could prevent S. aureus-induced proliferation of primary malignant T cells in Sézary syndrome PBMC cultures. Indeed, supernatant from S. aureus Pt - and recombinant SE induced comparable levels of mitotic (Ki67) and activated (CD25) primary malignant T cells (Fig. 6b). Importantly, endolysin treatment of S. aureus Pt supernatant prevented this effect (Fig. 6b). Similar results were obtained with PBMCs isolated at a later time-point from the same Sézary syndrome patient (Supl. fig. S2a). As endolysin strongly inhibited activation of non-malignant T cells (c.f. above), we examined whether endolysin-treatment also inhibited the effect of S. aureus Pt supernatant on proliferation of non-malignant T cells (Suppl. Fig. S2b and c). As expected, endolysin inhibited Ki67 expression in S. aureus Pt- treated patient-derived primary non-malignant CD4+ T cells (suppl. Fig. S2b) and non-malignant T cell lines (MyLa1850 cells - suppl Fig. S2c) supporting the hypothesis that endolysin inhibits S. aureus Pt mediated crosstalk between malignant and non-malignant T cells.
      Figure thumbnail gr6
      Figure 6Endolysin (XZ.700) inhibits S. aureus induced proliferation of malignant T cells. (a) Malignant T cell cell lines (SeAx and Myla3675) subjected to analysis of proliferation (Ki67 expression) using flow cytometry after culture for 24h with S. aureus Pt supernatant ± XZ.700 (S. aureus Pt sup and S. aureus Pt sup + XZ.700) in the presence or absence of non-malignant T cells (Myla1850). The level of Ki67 positive SeAx cell grown without S. aureus Pt supernatant in either mono-culture or in co-culture was comparable to the SeAx mono-culture experiments with 23.8 % ± 8.5 (STDEV) and 26 % ± 6.3 (STDEV) (n=3), respectively (data not shown). For Myla3675, background Ki67 positive cells were 27.5 % ± 2.3 (STDEV) and 37.4 % ± 4.1 (n=3), respectively (data not shown). Malignant and non-malignant T cell lines were distinguished by CellTrace Violet staining (b) PBMC isolated from a Sézary syndrome patient subjected to analysis of activation (CD25 expression) and proliferation (Ki67 expression) using flow cytometry after culture for three and six days. Cells were cultured in the presence of PBS as a negative control (Control), a pool of S. aureus enterotoxins (SEA SEB, SEC2, SED, SEI; SE-pool), supernatant from S. aureus Pt (S. aureus Pt sup), or S. aureus Pt supernatant with endolysin (S. aureus Pt sup +XZ.700). Malignant T cells were gated as CD3+, CD4+, CD7dim/-, and CD26-.
      In conclusion, these findings indicate that endolysin treatment profoundly reduced bacterial counts and SE-release to levels below the threshold required for induction of malignant T cell proliferation in co-culture with non-malignant cells.

      Discussion

      Here, we present evidence that recombinant endolysin XZ.700 has the potential to reduce S. aureus on the skin of CTCL patients. Thus, endolysin profoundly inhibited skin colonization by S. aureus of both healthy skin and lesional CTCL skin. Importantly, endolysin killed S. aureus after the S. aureus colonization had been established. This suggests that endolysin has potential in both a treatment-setting and as a prophylactic treatment. Although treatment with endolysin strongly inhibited S. aureus-mediated release of SE, the blockage was not complete. Importantly, only a low number of SE molecules per cell is needed to trigger a T cell response (
      • Herman A
      • Kappler JW
      • Marrack P
      • Pullen AM
      Superantigens: mechanism of T-cell stimulation and role in immune responses.
      ). Yet, endolysin-treated S. aureus Pt culture supernatants induced little or no T cell activation and proliferation indicating that endolysin-mediated reduction in S. aureus (and SE-secretion) was sufficient to prevent the mitogenic effect. Thus, our data show that endolysin profoundly inhibited both skin colonization by live patient-derived S. aureus and their ability to stimulate activation and proliferation of primary malignant T cells and T cell lines. In addition, we show that S. aureus Pt culture supernatants induced selective expression of IFNγ and IFNγ-inducible CXCL10 in healthy skin. Since CXCL10 is known to play a key role in CD4 T cell epidermotropism (
      • Sarris AH
      • Daliani D
      • Ulmer R
      • Crow M
      • Broxmeyer HE
      • Pugh W
      • et al.
      Interferon-inducible protein-10 and the pathogenesis of cutaneous T-cell lymphomas.
      ,
      • Sarris AH
      • Esgleyes-Ribot T
      • Crow M
      • Broxmeyer HE
      • Karasavvas N
      • Pugh W
      • et al.
      Cytokine loops involving interferon-gamma and IP-10, a cytokine chemotactic for CD4+ lymphocytes: an explanation for the epidermotropism of cutaneous T-cell lymphoma?.
      ,
      • Tensen CP
      • Vermeer MH
      • van der Stoop PM
      • van Beek P
      • Scheper RJ
      • Boorsma DM
      • et al.
      Epidermal interferon-gamma inducible protein-10 (IP-10) and monokine induced by gamma-interferon (Mig) but not IL-8 mRNA expression is associated with epidermotropism in cutaneous T cell lymphomas.
      ), we hypothesize that this could be a novel mechanism by which S. aureus modulates the TME and potentially promotes disease progresion and, consequently, another potential benefit from endolysin-mediated removal of S. aureus from lesional skin.
      Similar to antibiotics, resistance against bacteriophages exists. However, resistance against recombinant endolysins is less likely due to (i) their highly conserved targets in the PG and (ii) their action on the cell envelope, without having to enter the cell (when applied from the outside), thereby avoiding a majority of known resistance mechanisms (
      • Schmelcher M
      • Loessner MJ
      Bacteriophage endolysins - extending their application to tissues and the bloodstream.
      ). As recombinant endolysins have only been around for a few years, it is imposible to know whether S. aureus may eventually develop resistance to recombinant endolysin in a real life setting, but studies suggest that S. aureus do not develop resistance to endolysin-mediated killing in long-term in vitro cultures (
      • Eichenseher F
      • Herpers BL
      • Badoux P
      • Leyva-Castillo JM
      • Geha RS
      • van der Zwart M
      • et al.
      Linker-Improved Chimeric Endolysin Selectively Kills Staphylococcus aureus In Vitro, on Reconstituted Human Epidermis, and in a Murine Model of Skin Infection.
      ,
      • Kuiper JWP
      • Hogervorst JMA
      • Herpers BL
      • Bakker AD
      • Klein-Nulend J
      • Nolte PA
      • et al.
      The novel endolysin XZ.700 effectively treats MRSA biofilms in two biofilm models without showing toxicity on human bone cells in vitro.
      ). Moreover, XZ.700 was recently shown to be very efficient against MRSA and MRSA in biofilms (
      • Kuiper JWP
      • Hogervorst JMA
      • Herpers BL
      • Bakker AD
      • Klein-Nulend J
      • Nolte PA
      • et al.
      The novel endolysin XZ.700 effectively treats MRSA biofilms in two biofilm models without showing toxicity on human bone cells in vitro.
      ) (which are otherwise highly resistant to antibiotics (
      • Bjarnsholt T
      • Whiteley M
      • Rumbaugh KP
      • Stewart PS
      • Jensen PO
      • Frimodt-Moller N
      The importance of understanding the infectious microenvironment.
      )). This implies that endolysins like XZ.700 have a potential for topical eradication of various subtypes of S. aureus. Importantly, XZ.700 is not active against S. epidermidis (
      • Eichenseher F
      • Herpers BL
      • Badoux P
      • Leyva-Castillo JM
      • Geha RS
      • van der Zwart M
      • et al.
      Linker-Improved Chimeric Endolysin Selectively Kills Staphylococcus aureus In Vitro, on Reconstituted Human Epidermis, and in a Murine Model of Skin Infection.
      ). Whether the targeted killing of S. aureus by endolysin will be sufficient to reinforce beneficial commensal bacteria or enable other potential pathogenic bacteria to increase in presence will remain to be seen.
      Thus, studies are warranted to address whether recombinant endolysins are useful as a non-antibiotic alternative for eradication of S. aureus colonization from lesional skin and/or as prophylactic measures against skin colonization by S. aureus in CTCL patients. Since S. aureus also plays an important role in other dermatological diseases, we speculate that recombinant endolysin may also be of interest for anti-S. aureus treatment and prophylaxis against S. aureus-mediated flare-ups in diseases, such as, atopic dermatitis.
      In conclusion, we provide evidence that a recombinant endolysin, XZ.700, potently inhibits proliferation of pathogenic S. aureus, their skin colonization, and their tumor-promoting effects on malignant T cells. These findings suggest that recombinant endolysin comprises a non-antibiotic treatment strategy to control S. aureus colonization of lesional skin in CTCL.

      MATERIALS AND METHODS

      Planktonic S. aureus culture and endolysin activity assay

      S. aureus NCTC 8325, designated S. aureus LAB, and the clinical CTCL patient isolates Pt4.1.1.1 (S. aureus Pt), Pt6.1.1.1 (S. aureus Pt 1), and Pt7.1.1.1 (S. aureus Pt 2) were used (
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ). The clinical S. aureus isolates were identified by detection of coagulase and catalase activity and by MALDI-TOF MS. In addition, the isolates were characterized by multilocus sequence typing and characterized in relation to toxin genes by NGS as previously described (
      • Lindahl LM
      • Iversen L
      • Odum N
      • Kilian M
      Staphylococcus aureus and Antibiotics in Cutaneous T-Cell Lymphoma.
      ,
      • Lindahl LM
      • Willerslev-Olsen A
      • Gjerdrum LMR
      • Nielsen PR
      • Blumel E
      • Rittig AH
      • et al.
      Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
      ). S. aureus was prepared from an overnight culture diluted to an OD600nm of 0.01 (Shimadzu UV spectrophotometer, UV-1800) in tryptic soy broth (TSB) and regrown till early log phase (OD600nm of 0.2-0.7) at 37°C under shaking conditions (180 rpm). Colony forming units (CFU) was determined by serial dilution, plating on LB agar plates, overnight incubation at 37 °C, and counting the number of colonies. The growth inhibitory effect of XZ.700 (Micreos Pharma B.V., Netherlands) in liquid culture was determined by measuring the OD595m (Victor X4 2030 multiplate reader, PerkinElmer). XZ.700 was heat-inactivating at 70°C for 10 min.

      Patients skin specimens

      Biopsies from CTCL patients were obtained from donors at Bispebjerg Hospital and Gentofte Hospital, Denmark. In accordance with the Declaration of Helsinki, all biopsies were obtained with written, informed consent after approval by the Committee on Health Research Ethics (H-16025331). Healthy adult human skin designated “healthy skin” were obtained from donors below the age of 70 undergoing blepharoplasty at ophthalmologist Neel Gerner, Lyngby, Denmark. The use of anonymized leftover skin from cosmetic surgeries was obtained with written, informed consent and approved by the Danish Data Protection Agency. Healthy skin biopsies and CTCL patient biopsies were maintained in DMEM (Thermo, #11965) and Ham’s F12 (Thermo, #11765) supplemented with 2% heat-inactivated FBS (Biological Industries, #04-007-1A), EGF (Sigma, #SRP3027), insulin (Merck, #I9278), and penicillin/streptomycin (Sigma-Aldrich, #P7539). Biopsies were incubated in a humidified 37 °C chamber with 5% CO2.

      Ex vivo skin experiments

      S. aureus (OD600nm 0.2-0.7) was resuspended to about 1·106 CFU/10 μL PBS based on previous correlation between OD600nm and CFU. Actual CFU seeded was determined for each seeding. S. aureus was seeded at a density of about 5·106 CFU/cm2 (1-10·106 CFU/cm2). Following incubation, biopsies were washed in 0.1 M citrate buffer (pH 4.5) and S. aureus was released by swabbing with a cotton bud 10 times, transferred to 0.1 M citrate buffer (pH 4.5) by sonication (5 min degassing, 5 min sonication (Branson 2510)) and 10 sec vortex before determining CFU.

      Immunofluorescence labeling and imaging

      Skin biopsies were fixed in fresh 4% paraformaldehyde at 4 °C and paraffin embedded. 3-5μm sections were used for hematoxylin and eosin staining (Histolab, Department of Biomedical Sciences, University of Copenhagen) or fluorescence in situ hybridization (FISH) after deparaffinization. FISH was performed using fluorescently labeled PNA-probes for S. aureus (AdvanDx, #Sta16S03-TXR, OpGen) according to manufacturer’s protocol, stained with DAPI (Thermo, #62248) and mounted with ProLong Diamont Antifade Reagent (Thermo Fisher, #P36961). Images were collected using Zeiss Axio Scan.Z1 with a 405 nm (Dapi) and a 561 nm (Texas red) laser lines and a 10x/0.45 objective.

      S. aureus supernatants and S. aureus enterotoxin (SE) determination

      S. aureus was prepared from an overnight culture diluted to an OD600nm of 0.01 (Shimadzu UV spectrophotometer, UV-1800) in TSB) and regrown for 4 hours with or without 1μg/mL XZ.700. S. aureus Pt supernatant was prepared by centrifugation (10.000xg for 10 min) followed by sterile filtration (0.22μm filter). SE in S. aureus supernatants were detected by ELISA according to manufacturer’s protocol; SEA (Chrondrex, #6029), SE (R-Biopharm, RIDASCREEN SET A/B/C/D/E kit). Signal was detected by UV plate reader (Thermoscientific, Multiscan FC).

      Cell lines

      The T cell lines SeAx (
      • Kaltoft K
      • Bisballe S
      • Rasmussen HF
      • Thestrup-Pedersen K
      • Thomsen K
      • Sterry W
      A continuous T-cell line from a patient with Sezary syndrome.
      ), MyLa1850 (
      • Kaltoft K
      • Thestrup-Pedersen K
      • Jensen JR
      • Bisballe S
      • Zachariae H
      Establishment of T and B cell lines from patients with mycosis fungoides.
      ), and MyLa3675 (
      • Krejsgaard T
      • Vetter-Kauczok CS
      • Woetmann A
      • Lovato P
      • Labuda T
      • Eriksen KW
      • et al.
      Jak3- and JNK-dependent vascular endothelial growth factor expression in cutaneous T-cell lymphoma.
      ) were cultured in RPMI1640 (Sigma, #R2405) containing penicillin/streptomycin (Sigma-Aldrich, #P7539), 10% human serum (Bloodbank, Copenhagen University Hospital, Denmark), IL-2, and for MyLa1850 also IL-4 as described (
      • Woetmann A
      • Lovato P
      • Eriksen KW
      • Krejsgaard T
      • Labuda T
      • Zhang Q
      • et al.
      Nonmalignant T cells stimulate growth of T-cell lymphoma cells in the presence of bacterial toxins.
      ).

      Sézary Syndrome patient PBMC culture and Flow cytometry

      Peripheral blood mononuclear cells (PBMCs) were isolated from Sézary Syndrome patient blood by lymphoprep density-gradient centrifugation (Stemcell Technologies, Vancouver, Canada). SE (50 ng/mL; SEA, SEB, SEC2, SED, SEI; Toxin Technology, Sarasota, FL, USA). Cells were stained for CellTrace, propidium iodide and Fixable Viability Stain Dye eFluor780 (ThermoFisher) and fluorochrome-conjugated CD3, CD4, CD7, CD8, CD19, CD25, CD26, Ki-67, pY-STAT5 and respective fluorochrome-conjugated isotype control antibodies (Biolegend,San Diego, CA, USA; BD Biosciences, San Jose, CA, USA). Flow cytometry analysis was performed on a LSR Fortessa flow cytometer (BD Biosciences) using FlowJo software (Tree Star, Ashland, OR). Malignant T cells were gated as CD3+ CD4+, CD7dim/-, and CD26-. Non-malignant T cells were gated as CD3+ CD4+, CD7+, and CD26+.

      Quantitative reverse transcription PCR (RT-qPCR)

      Biopsies were homogenized (Tissuelyser at 20000 HZ, Qiagen or Precellys, hard tissue program, Bertin) prior to RNA isolation (Qiagen #74134 or AllPrep DNA/RNA kit). cDNA was prepared (Applied biosystems, #4368814) and subjected to RT-qPCR (Lightcycler480) using β-actin as reference gene (TaqMan probe, ThermoFisher). Relative expression was calculated according to the 2ΔΔCT method.
      Statistics
      All statistical analyses and graphs were done using GraphPad Prism® software (Prism 9.0.1 version). For statistical analysis, a 2-tailed Student’s t test or an Ordinary one-way ANOVA with Dunnett's multiple comparisons test were made. * significant at p<0.05, ** significant at p>0.01, *** significant at p<0.001, **** significant at p<0.0001. Error bars represent standard deviation.

      DATA AVAILABILITY STATEMENT

      No datasets were generated or analyzed during the current study.

      CONFLICT OF INTERESTS

      Niels Ødum has an advisory consultant honorarium from Micreos Pharma B.V and Almirall. Bob de Rooij and Johan Frieling work for Micreos Pharma B.V., Christian Röhrig, and Mathias Schmelcher work for Micreos GmbH. Lars Iversen served as a consultant and/or paid speaker for and/or participated in clinical trials sponsored by AbbVie, Almirall, Amgen, Astra Zeneca, BMS, Boehringer Ingelheim, Celgene, Centocor, Eli Lilly, Janssen Cilag, Kyowa, Leo Pharma, Micreos Human Health, MSD, Novartis, Pfizer, Regranion, Samsung, Union Therapeutics, and UCB. Koralov Laboratory (Sergei Koralov) has a sponsored research agreement with Micreos company to explore potential for recombinant endolysins as a de-colonization agent in the context of T cell lymphoma. This sponsored work is not covered by the present manuscript and the agreement did not influence interpretation of results. All other authors declare no potential conflicts of interest.

      ACKNOWLEDGEMENTS

      This research was funded by LEO Foundation, The Danish Cancer Society (Kræftens Bekæmpelse), the Fight Cancer Program (Knæk Cancer), Novo Nordisk Research Foundation, Novo Nordisk Foundation Tandem Program (grant number NNF21OC0066950), and The Danish Council for Independent Research (Danmarks Frie Forskningsfond, 2 project grants).

      Supplementary Material

      References

        • Allam G
        • Gaber AM
        • Othman SI
        • Abdel-Moneim A
        The potential role of interleukin-37 in infectious diseases.
        Int Rev Immunol. 2020; 39: 3-10
        • Aschebrook-Kilfoy B
        • Cocco P
        • La Vecchia C
        • Chang ET
        • Vajdic CM
        • Kadin ME
        • et al.
        Medical history, lifestyle, family history, and occupational risk factors for mycosis fungoides and Sezary syndrome: the InterLymph Non-Hodgkin Lymphoma Subtypes Project.
        J Natl Cancer Inst Monogr. 2014; 2014: 98-105
        • Axelrod PI
        • Lorber B
        • Vonderheid EC
        Infections complicating mycosis fungoides and Sezary syndrome.
        JAMA : the journal of the American Medical Association. 1992; 267: 1354-1358
        • Bagot M
        • Nikolova M
        • Schirm-Chabanette F
        • Wechsler J
        • Boumsell L
        • Bensussan A
        Crosstalk between tumor T lymphocytes and reactive T lymphocytes in cutaneous T cell lymphomas.
        Ann N Y Acad Sci. 2001; 941: 31-38
        • Bjarnsholt T
        • Whiteley M
        • Rumbaugh KP
        • Stewart PS
        • Jensen PO
        • Frimodt-Moller N
        The importance of understanding the infectious microenvironment.
        Lancet Infect Dis. 2022; 22: e88-e92
        • Blumel E
        • Munir Ahmad S
        • Nastasi C
        • Willerslev-Olsen A
        • Gluud M
        • Fredholm S
        • et al.
        Staphylococcus aureus alpha-toxin inhibits CD8(+) T cell-mediated killing of cancer cells in cutaneous T-cell lymphoma.
        Oncoimmunology. 2020; 91751561
        • Blumel E
        • Willerslev-Olsen A
        • Gluud M
        • Lindahl LM
        • Fredholm S
        • Nastasi C
        • et al.
        Staphylococcal alpha-toxin tilts the balance between malignant and non-malignant CD4(+) T cells in cutaneous T-cell lymphoma.
        Oncoimmunology. 2019; 8e1641387
        • de Masson A
        • Darbord D
        • Dobos G
        • Boisson M
        • Roelens M
        • Ram-Wolff C
        • et al.
        Macrophage-derived CXCL9 and CXCL11, T-cell skin homing, and disease control in mogamulizumab-treated CTCL patients.
        Blood. 2022; 139: 1820-1832
        • Dobos G
        • Calugareanu A
        • Michel L
        • Battistella M
        • Ram-Wolff C
        • Bouaziz JD
        • et al.
        Exploring the role of the skin microenvironment in cutaneous T-cell lymphoma using single cell RNA-sequencing.
        Eur J Cancer. 2021; 156: S3-S4
        • Dobos G
        • de Masson A
        • Ram-Wolff C
        • Beylot-Barry M
        • Pham-Ledard A
        • Ortonne N
        • et al.
        Epidemiological changes in cutaneous lymphomas: an analysis of 8593 patients from the French Cutaneous Lymphoma Registry.
        The British journal of dermatology. 2021; 184: 1059-1067
        • Dummer R
        • Vermeer MH
        • Scarisbrick JJ
        • Kim YH
        • Stonesifer C
        • Tensen CP
        • et al.
        Cutaneous T cell lymphoma.
        Nat Rev Dis Primers. 2021; 7: 61
        • Edelson RL
        Cutaneous T cell lymphoma: mycosis fungoides, Sezary syndrome, and other variants.
        Journal of the American Academy of Dermatology. 1980; 2: 89-106
        • Edelson RL
        Cutaneous T cell lymphoma: the helping hand of dendritic cells.
        Ann N Y Acad Sci. 2001; 941: 1-11
        • Eichenseher F
        • Herpers BL
        • Badoux P
        • Leyva-Castillo JM
        • Geha RS
        • van der Zwart M
        • et al.
        Linker-Improved Chimeric Endolysin Selectively Kills Staphylococcus aureus In Vitro, on Reconstituted Human Epidermis, and in a Murine Model of Skin Infection.
        Antimicrob Agents Chemother. 2022; e0227321
        • Emge DA
        • Bassett RL
        • Duvic M
        • Huen AO
        Methicillin-resistant Staphylococcus aureus (MRSA) is an important pathogen in erythrodermic cutaneous T-cell lymphoma (CTCL) patients.
        Arch Dermatol Res. 2020; 312: 283-288
        • Fanok MH
        • Sun A
        • Fogli LK
        • Narendran V
        • Eckstein M
        • Kannan K
        • et al.
        Role of Dysregulated Cytokine Signaling and Bacterial Triggers in the Pathogenesis of Cutaneous T-Cell Lymphoma.
        J Invest Dermatol. 2018; 138: 1116-1125
        • Geskin LJ
        • Akilov OE
        • Lin Y
        • Lokshin AE
        Distinct age-matched serum biomarker profiles in patients with cutaneous T-cell lymphoma.
        Exp Dermatol. 2014; 23: 598-600
        • Gluud M
        • Pallesen EMH
        • Buus TB
        • Gjerdrum LMR
        • Lindahl LM
        • Kamstrup MR
        • et al.
        Malignant T cells induce skin barrier defects through cytokine-mediated JAK/STAT signalling in cutaneous T-cell lymphoma.
        Blood. 2022;
        • Gluud M
        • Willerslev-Olsen A
        • Gjerdrum LMR
        • Lindahl LM
        • Buus TB
        • Andersen MH
        • et al.
        MicroRNAs in the Pathogenesis, Diagnosis, Prognosis and Targeted Treatment of Cutaneous T-Cell Lymphomas.
        Cancers (Basel). 2020; 12
        • Herman A
        • Kappler JW
        • Marrack P
        • Pullen AM
        Superantigens: mechanism of T-cell stimulation and role in immune responses.
        Annu Rev Immunol. 1991; 9: 745-772
        • Herrera A
        • Cheng A
        • Mimitou EP
        • Seffens A
        • George D
        • Bar-Natan M
        • et al.
        Multimodal single-cell analysis of cutaneous T-cell lymphoma reveals distinct subclonal tissue-dependent signatures.
        Blood. 2021; 138: 1456-1464
        • Hodak E
        • Amitay-Laish I
        Mycosis fungoides: A great imitator.
        Clin Dermatol. 2019; 37: 255-267
        • Jackow CM
        • Cather JC
        • Hearne V
        • Asano AT
        • Musser JM
        • Duvic M
        Association of erythrodermic cutaneous T-cell lymphoma, superantigen-positive Staphylococcus aureus, and oligoclonal T-cell receptor V beta gene expansion.
        Blood. 1997; 89: 32-40
        • Kadin ME
        • Hamilton RG
        • Vonderheid EC
        Evidence linking atopy and staphylococcal superantigens to the pathogenesis of lymphomatoid papulosis, a recurrent CD30+ cutaneous lymphoproliferative disorder.
        PLoS One. 2020; 15e0228751
        • Kaltoft K
        • Bisballe S
        • Rasmussen HF
        • Thestrup-Pedersen K
        • Thomsen K
        • Sterry W
        A continuous T-cell line from a patient with Sezary syndrome.
        Arch Dermatol Res. 1987; 279: 293-298
        • Kaltoft K
        • Thestrup-Pedersen K
        • Jensen JR
        • Bisballe S
        • Zachariae H
        Establishment of T and B cell lines from patients with mycosis fungoides.
        The British journal of dermatology. 1984; 111: 303-308
        • Kim YH
        • Bagot M
        • Pinter-Brown L
        • Rook AH
        • Porcu P
        • Horwitz SM
        • et al.
        Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): an international, open-label, randomised, controlled phase 3 trial.
        Lancet Oncol. 2018; 19: 1192-1204
        • Krejsgaard T
        • Lindahl LM
        • Mongan NP
        • Wasik MA
        • Litvinov IV
        • Iversen L
        • et al.
        Malignant inflammation in cutaneous T-cell lymphoma-a hostile takeover.
        Semin Immunopathol. 2017; 39: 269-282
        • Krejsgaard T
        • Litvinov IV
        • Wang Y
        • Xia L
        • Willerslev-Olsen A
        • Koralov SB
        • et al.
        Elucidating the role of interleukin-17F in cutaneous T-cell lymphoma.
        Blood. 2013; 122: 943-950
        • Krejsgaard T
        • Vetter-Kauczok CS
        • Woetmann A
        • Lovato P
        • Labuda T
        • Eriksen KW
        • et al.
        Jak3- and JNK-dependent vascular endothelial growth factor expression in cutaneous T-cell lymphoma.
        Leukemia. 2006; 20: 1759-1766
        • Kuiper JWP
        • Hogervorst JMA
        • Herpers BL
        • Bakker AD
        • Klein-Nulend J
        • Nolte PA
        • et al.
        The novel endolysin XZ.700 effectively treats MRSA biofilms in two biofilm models without showing toxicity on human bone cells in vitro.
        Biofouling. 2021; 37: 184-193
        • Lewis DJ
        • Holder BB
        • Duvic M
        The "Duvic regimen" for erythrodermic flares secondary to Staphylococcus aureus in mycosis fungoides and Sezary syndrome.
        Int J Dermatol. 2018; 57: 123-124
        • Lindahl LM
        • Iversen L
        • Odum N
        • Kilian M
        Staphylococcus aureus and Antibiotics in Cutaneous T-Cell Lymphoma.
        Dermatology. 2021; : 1-3
        • Lindahl LM
        • Willerslev-Olsen A
        • Gjerdrum LMR
        • Nielsen PR
        • Blumel E
        • Rittig AH
        • et al.
        Antibiotics inhibit tumor and disease activity in cutaneous T cell lymphoma.
        Blood. 2019;
        • Mehdi SJ
        • Moerman-Herzog A
        • Wong HK
        Normal and cancer fibroblasts differentially regulate TWIST1, TOX and cytokine gene expression in cutaneous T-cell lymphoma.
        BMC Cancer. 2021; 21: 492
        • Mirvish ED
        • Pomerantz RG
        • Geskin LJ
        Infectious agents in cutaneous T-cell lymphoma.
        Journal of the American Academy of Dermatology. 2011; 64: 423-431
        • Qu K
        • Zaba LC
        • Satpathy AT
        • Giresi PG
        • Li R
        • Jin Y
        • et al.
        Chromatin Accessibility Landscape of Cutaneous T Cell Lymphoma and Dynamic Response to HDAC Inhibitors.
        Cancer Cell. 2017; 32: 27-41 e4
        • Sarris AH
        • Daliani D
        • Ulmer R
        • Crow M
        • Broxmeyer HE
        • Pugh W
        • et al.
        Interferon-inducible protein-10 and the pathogenesis of cutaneous T-cell lymphomas.
        Leuk Lymphoma. 1996; 24: 103-110
        • Sarris AH
        • Esgleyes-Ribot T
        • Crow M
        • Broxmeyer HE
        • Karasavvas N
        • Pugh W
        • et al.
        Cytokine loops involving interferon-gamma and IP-10, a cytokine chemotactic for CD4+ lymphocytes: an explanation for the epidermotropism of cutaneous T-cell lymphoma?.
        Blood. 1995; 86: 651-658
        • Schmelcher M
        • Donovan DM
        • Loessner MJ
        Bacteriophage endolysins as novel antimicrobials.
        Future Microbiol. 2012; 7: 1147-1171
        • Schmelcher M
        • Loessner MJ
        Bacteriophage endolysins - extending their application to tissues and the bloodstream.
        Curr Opin Biotechnol. 2021; 68: 51-59
        • Schmelcher M
        • Shen Y
        • Nelson DC
        • Eugster MR
        • Eichenseher F
        • Hanke DC
        • et al.
        Evolutionarily distinct bacteriophage endolysins featuring conserved peptidoglycan cleavage sites protect mice from MRSA infection.
        J Antimicrob Chemother. 2015; 70: 1453-1465
        • Stolearenco V
        • Namini MRJ
        • Hasselager SS
        • Gluud M
        • Buus TB
        • Willerslev-Olsen A
        • et al.
        Cellular Interactions and Inflammation in the Pathogenesis of Cutaneous T-Cell Lymphoma.
        Front Cell Dev Biol. 2020; 8: 851
        • Talpur R
        • Bassett R
        • Duvic M
        Prevalence and treatment of Staphylococcus aureus colonization in patients with mycosis fungoides and Sezary syndrome.
        The British journal of dermatology. 2008; 159: 105-112
        • Tensen CP
        • Quint KD
        • Vermeer MH
        Genetic and epigenetic insights into cutaneous T-cell lymphoma.
        Blood. 2022; 139: 15-33
        • Tensen CP
        • Vermeer MH
        • van der Stoop PM
        • van Beek P
        • Scheper RJ
        • Boorsma DM
        • et al.
        Epidermal interferon-gamma inducible protein-10 (IP-10) and monokine induced by gamma-interferon (Mig) but not IL-8 mRNA expression is associated with epidermotropism in cutaneous T cell lymphomas.
        J Invest Dermatol. 1998; 111: 222-226
        • Tokura Y
        • Heald PW
        • Yan SL
        • Edelson RL
        Stimulation of cutaneous T-cell lymphoma cells with superantigenic staphylococcal toxins.
        J Invest Dermatol. 1992; 98: 33-37
        • Tokura Y
        • Yagi H
        • Ohshima A
        • Kurokawa S
        • Wakita H
        • Yokote R
        • et al.
        Cutaneous colonization with staphylococci influences the disease activity of Sezary syndrome: a potential role for bacterial superantigens.
        The British journal of dermatology. 1995; 133: 6-12
        • van Doorn R
        • Slieker RC
        • Boonk SE
        • Zoutman WH
        • Goeman JJ
        • Bagot M
        • et al.
        Epigenomic Analysis of Sezary Syndrome Defines Patterns of Aberrant DNA Methylation and Identifies Diagnostic Markers.
        J Invest Dermatol. 2016; 136: 1876-1884
        • Vieyra-Garcia P
        • Crouch JD
        • O'Malley JT
        • Seger EW
        • Yang CH
        • Teague JE
        • et al.
        Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma.
        JCI Insight. 2019; 4
        • Willerslev-Olsen A
        • Buus TB
        • Nastasi C
        • Blumel E
        • Gluud M
        • Bonefeld CM
        • et al.
        Staphylococcus aureus enterotoxins induce FOXP3 in neoplastic T cells in Sezary syndrome.
        Blood cancer journal. 2020; 10: 57
        • Willerslev-Olsen A
        • Gjerdrum LMR
        • Lindahl LM
        • Buus TB
        • Pallesen EMH
        • Gluud M
        • et al.
        Staphylococcus aureus Induces Signal Transducer and Activator of Transcription 5Dependent miR-155 Expression in Cutaneous T-Cell Lymphoma.
        J Invest Dermatol. 2021; 141: 2449-2458
        • Willerslev-Olsen A
        • Krejsgaard T
        • Lindahl LM
        • Bonefeld CM
        • Wasik MA
        • Koralov SB
        • et al.
        Bacterial toxins fuel disease progression in cutaneous T-cell lymphoma.
        Toxins (Basel). 2013; 5: 1402-1421
        • Willerslev-Olsen A
        • Krejsgaard T
        • Lindahl LM
        • Litvinov IV
        • Fredholm S
        • Petersen DL
        • et al.
        Staphylococcal enterotoxin A (SEA) stimulates STAT3 activation and IL-17 expression in cutaneous T-cell lymphoma.
        Blood. 2016; 127: 1287-1296
        • Woetmann A
        • Lovato P
        • Eriksen KW
        • Krejsgaard T
        • Labuda T
        • Zhang Q
        • et al.
        Nonmalignant T cells stimulate growth of T-cell lymphoma cells in the presence of bacterial toxins.
        Blood. 2007; 109: 3325-3332
        • Wu X
        • Singh R
        • Hsu DK
        • Zhou Y
        • Yu S
        • Han D
        • et al.
        A Small Molecule CCR2 Antagonist Depletes Tumor Macrophages and Synergizes with Anti-PD-1 in a Murine Model of Cutaneous T-Cell Lymphoma (CTCL).
        J Invest Dermatol. 2020; 140: 1390-13400 e4
        • Wu XS
        • Lonsdorf AS
        • Hwang ST
        Cutaneous T-cell lymphoma: roles for chemokines and chemokine receptors.
        J Invest Dermatol. 2009; 129: 1115-1119
        • Yu KK
        • Smith NP
        • Essien SV
        • Teague JE
        • Vieyra-Garcia P
        • Gehad A
        • et al.
        IL-32 supports the survival of malignant T cells in cutaneous T cell lymphoma.
        J Invest Dermatol. 2022;