You are on page 1of 16

MOLECULAR CANCER THERAPEUTICS | LARGE MOLECULE THERAPEUTICS

Brentuximab Vedotin–Driven Microtubule Disruption


Results in Endoplasmic Reticulum Stress Leading to
Immunogenic Cell Death and Antitumor Immunity
Ryan A. Heiser, Anthony T. Cao, Weiping Zeng, Michelle Ulrich, Patrick Younan, Martha E. Anderson,
Esther S. Trueblood, Mechthild Jonas, Robert Thurman, Che-Leung Law, and Shyra J. Gardai

ABSTRACT

Brentuximab vedotin, a CD30-directed antibody–drug conju- from tumor rechallenge; in addition, T cells transferred from
gate (ADC), is approved for clinical use in multiple CD30-expres- previously vaccinated animals slowed tumor growth in immu-
sing lymphomas. The cytotoxic payload component of brentux- nodeficient mice. Immunity acquired from killed tumor cell
imab vedotin is monomethyl auristatin E (MMAE), a highly potent vaccination was further amplified by the addition of PD-1
microtubule-disrupting agent. Preclinical results provided here blockade. In a humanized model of CD30þ B-cell tumors,
demonstrate that treatment of cancer cells with brentuximab treatment with brentuximab vedotin drove the expansion and

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


vedotin or free MMAE leads to a catastrophic disruption of the recruitment of autologous Epstein-Barr virus–reactive CD8þ T
microtubule network eliciting a robust endoplasmic reticulum cells potentiating the activity of anti–PD-1 therapy. Together,
(ER) stress response that culminates in the induction of the classic these data support the ability of brentuximab vedotin and
hallmarks of immunogenic cell death (ICD). In accordance with MMAE to drive ICD in tumor cells resulting in the activation
the induction of ICD, brentuximab vedotin–killed lymphoma cells of antigen-presenting cells and augmented T-cell immunity.
drove innate immune cell activation in vitro and in vivo. In the These data provide a strong rationale for the clinical combina-
“gold-standard” test of ICD, vaccination of mice with brentuximab tion of brentuximab vedotin and other MMAE-based ADCs with
vedotin or free MMAE-killed tumor cells protected animals checkpoint inhibitors.

Introduction cellular environment promotes APC recruitment via purinergic


receptors and activation via the nucleotide-binding domain, leu-
Tumors evade immune surveillance through tolerance of tumor-
cine-rich–containing family, pyrin domain–containing-3 (NLRP3)
associated neoantigens or by evolving immunosuppressive tumor
inflammasome. Finally, HMGB1 release directly activates APCs
microenvironments (TME; refs. 1, 2). Therapeutics that kill tumor
via Toll-like receptor 4 (TLR4; refs. 6–11). Together, the spatio-
cells directly while also promoting tumor-specific T-cell responses may
temporal presentation of these damage-associated molecular pat-
potentiate checkpoint inhibitor therapies, and therefore promote
terns (DAMP), concomitant with tumor antigen exposure from
durable clinical responses (3).
dying tumor cells, can result in the development of antigen-specific
Apoptotic cell death is a normal aspect of cellular turnover and is
antitumor immunity (5).
associated with silent or tolerogenic immune responses (4). In
Although a variety of anticancer therapeutic approaches with
contrast, immunogenic cell death (ICD) is an inflammatory cell
different molecular mechanisms of action have been described pre-
death process that promotes the activation and recruitment of innate
viously to elicit ICD (radiotherapy, photodynamic therapy, anthra-
immune cells (1). During the pre-apoptotic stage of ICD, cells
cyclines, and oxaliplatin; refs. 5, 12, 13), a common factor is the
present a defined combination of hallmark signals that provoke a
dysregulation of ER homeostasis (1, 14–18). Overloading the ER’s
localized immune reaction to the dying cells (5). Cardinal hallmarks
capacity for unfolded polypeptides, or other disruptions to the protein-
of ICD include heightened translocation of calreticulin (CRT) from
folding environment, initiates a canonical ER stress response known as
the endoplasmic reticulum (ER) to the cell surface, active secretion of
the unfolded protein response (UPR). A strong UPR, coupled with the
ATP, and passive release of non-histone chromatin-binding protein
associated release of ER Ca2þ stores, is required to trigger ICD-related
high-mobility group box 1 (HMGB1; Supplementary Fig. S1; ref. 6).
hallmark DAMPs (7, 16, 17, 19). Translocation of CRT to the cell
CRT exposure promotes phagocytosis of dying cells by antigen-
surface and active secretion of ATP resulting from cellular autophagy
presenting cells (APC) via interactions with low-density lipoprotein
follow severe ER stress (7, 20–22), whereas the release of nuclear
receptor-related protein 1. In addition, ATP release into the extra-
HMGB1 is thought to occur as the dying cell loses nuclear and
plasma membrane integrity (22–24). Synthetic knockdown of the
ER stress response during treatment with ICD-inducing therapeu-
Seagen Inc., Bothell, Washington. tics abrogates ICD (21, 25). Conversely, combining pharmacolog-
Corresponding Author: Ryan A. Heiser, Seagen Inc., 21823 30th Drive S.E,
ical agents that perturb ER function with certain non–ICD-inducing
Bothell, WA, 98021. E-mail: rheiser@seagen.com compounds have been shown to increase the immunogenicity of
dying tumor cells (15, 26–28).
Mol Cancer Ther 2024;23:68–83
Although some systemic chemotherapies elicit ICD, they can also be
doi: 10.1158/1535-7163.MCT-23-0118
associated with lymphopenia and systemic immune alterations often
This open access article is distributed under the Creative Commons Attribution- requiring months for T cells to return to pre-treatment levels (29–32).
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license. Antibody–drug conjugates (ADC) are a clinically validated strategy for
2023 The Authors; Published by the American Association for Cancer Research the selected delivery of cytotoxic molecules to tumors that largely avoid

AACRJournals.org | 68
Brentuximab Vedotin and Immunogenic Cell Death

persistent depletion of peripheral lymphocytes (33). The combination Cell lines


of ICD-inducing ADCs with checkpoint inhibitors may therefore drive Cell lines L540 (DSMZ, ACC72; RRID:CVCL_1362; human
complementary immune mechanisms. Multiple combinations of female), KARPAS 299 (DSMZ, ACC31, RRID:CVCL_1324; human
ADCs with checkpoint inhibitors are being actively evaluated in the male), HDLM-2 (DSMZ, ACC17; RRID:CVCL_0009; human male),
clinic with promising early results (34, 35). and A20 (ATCC, TIB-208; RRID:CVCL_1940; mouse) were originally
Multiple preclinical studies have described successful combina- purchased from the vendors listed and since maintained by the Seagen
tions of ADCs with checkpoint inhibitors and other immunothera- cell bank (2016–2023). For each study, cell lines were requested from
pies. M€ uller and colleagues (36) were the first to show that the the cell bank and aliquots were thawed and cultured for periods
HER2-directed ADC trastuzumab emtansine (T-DM1) drove between 1 and 3 weeks. Cell bank cell lines are routinely checked
immune infiltration in breast cancer lesions in the clinic. In for contamination, including Mycoplasma, and cell line identity
addition, T-DM1 combined with cytotoxic T-lymphocyte antigen 4 authentication by an STR-based DNA profiling and multiplex PCR,
(CTLA-4) and programmed cell death protein 1 (PD-1) blockade CellCheck 16–Human, or CellCheck 19 Mouse Plus, IDEXX Labora-
amplified antitumor responses in preclinical models (36). Data from tories, Inc. Following any cell line modifications or cloning, expanded
this group supported the direct activation of APCs by dolastatins, lines were resubmitted for pathogen burden and cell line identity
from which monomethyl auristatin E (MMAE) was synthetically validation before experimental use.
derived, and also MMAE itself (37, 38). Rios-Doria and collea-
gues (39) showed that tubulysin and pyrrolobenzodiazepine dimer ICD hallmark assessment
(PBD)-based ADCs successfully combined with immunotherapies CRT exposure

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


in syngeneic tumor models, supportive of ICD induction. Finally, The lymphoma cell line L540cy was grown in RPMI and 20% FBS
monomethyl auristatin F, a derivative of MMAE with greatly (Gibco). Cells were treated with IC50 or IC90 concentrations of
reduced membrane permeability, was shown to drive ICD with the brentuximab vedotin, doxorubicin, etoposide, or oxaliplatin for 0,
B-cell maturation antigen-directed ADC belantamab mafodotin 18, 24, or 48 hours. Cells were also treated with controls of a non-
and combine with a checkpoint modulator (40). Together, these binding ADC (IgG-MMAE), chimeric anti-CD30 antibody (cAC10)
studies highlight the potential immunomodulatory abilities of ADC naked mAb at the IC50 or IC90 concentrations of brentuximab vedotin.
payloads, particularly those causing microtubule destabilization. Cells were harvested and stained for cell-surface CRT (FMC75, Enzo,
Indeed, results supporting ICD induction from vinca domain- RRID:AB_11180758). Staining was concomitant with propidium
binding microtubule-destabilizing agents were predicted by early iodide (PI; Sigma) and all staining was performed as per the manu-
ICD-screening platforms (41, 42). facturer’s recommendations. Surface levels of CRT on PI-negative cells
Brentuximab vedotin is a CD30-directed vedotin ADC, current- were analyzed by flow cytometry.
ly approved for use in the US for multiple CD30-expressing
lymphomas (43). Brentuximab vedotin comprises a CD30-directed ATP and HMGB1 release assays
monoclonal antibody (mAb) conjugated to a protease cleavable L540cy cells were plated at 200,000–3,000,000 cells/well on a 24-well
maleimidocaproyl–valine-citrulline (mc-vc) linker and the micro- dish and allowed to reach 50% confluence. Cells were then treated with
tubule-disrupting agent (MMAE; refs. 44, 45). Upon internalization, IC90 (1 mg/mL) of brentuximab vedotin or a non-binding ADC (IgG-
the protease-cleavable linker enables the preferential release of MMAE), 100 nmol/L (IC90) MMAE, 1 mg/mL oxaliplatin, or 30 mmol/L
MMAE within CD30-expressing target cells, resulting in microtu- etoposide for 18 hours. For additional cell lines shown in the Sup-
bule disruption and cell death (45–48). We previously reported on plementary Figures, approximately 50,000 cells were incubated with
the ability of brentuximab vedotin and MMAE to induce ICD in IC20–IC50 concentrations of test articles for 48–72 hours. After treat-
target cells via microtubule disruption and ER stress in a series of ment of cells, 250 mL of media were harvested from each well by tilting
in vitro and in vivo experiments (49, 50). Furthermore, the potential the plate and transferred to a 1.5-mL Eppendorf tube. Cells were spun
for brentuximab vedotin or MMAE treatment to activate antitumor at 1,600 rpm (200 rcf) for 1 minute in a microcentrifuge. Subsequently,
immunity in combination with PD-1 inhibition was previously 50 mL of media were transferred to wells in a 96-well clear-bottom plate
evaluated (51). Here, we expand upon our initial findings of and 50 mL of CellTiter Glo (Promega) was added to each well and
brentuximab vedotin and MMAE-induced ICD and the enhanced shielded from light. Plates were shaken for 1 minute on an orbital
immune response observed in combination with PD-1 inhibition. shaker, incubated for an additional 10 minutes at room temperature to
stabilize luminescent signal, and immediately read on an Envision
Plate Reader. ATP levels were normalized to levels from untreated cells.
Materials and Methods HMGB1 release was assessed in a similar manner to ATP release.
Animals and animal care L540cy cells were plated and treated for 18–72 hours. Extracellular
All animal studies were executed in compliance with institutional HMBG1 was measured in supernatants using an HMGB1 ELISA
guidelines and regulations and approved by an Institutional Animal (Tecan, IBL International).
Care and Use Committee (IACUC) and internal review committee
under protocols SGE-024 and SGE-029. Mice were age- and sex- Fluorescent staining
matched and randomly assigned to treatment groups in all experi- An Enzo Life Sciences’ ER-ID kit was used for fluorescent labeling of
ments with females being predominantly used for studies. The number the ER in L540cy cells. Antibodies against phosphorylated inositol–
of mice used in each experiment was determined on the basis of requiring enzyme 1 (pIRE1; NB100–2323, Novus Biologicals, RRID:
previous experience with variability of model performance. Mice were AB_0145203) and anti-tubulin (YOL1/34, Abcam, RRID:AB_305329),
housed in an IACUC-certified housing facility under the supervision of were used for fluorescent imaging. L540cy cell nuclei were stained using
a licensed veterinarian. All animal studies were designed to minimize Diamond Antifade Mountant with 40 ,6-diamidino-2-phenylindole
animal usage and distress. (Thermo Fisher Scientific).

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 69


Heiser et al.

ER stress determination by western blot analysis treatment, tumors were harvested, weighed, and manually dissoci-
L540cy cells were plated at 500,000 cells/well in a 12-well plate ated through a 70-mm cell strainer to generate single-cell suspen-
overnight. Cells were then treated with IC90 (1 mg/mL) of brentuximab sions for flow cytometry. Tumor cell suspensions were stained with
vedotin or a non-binding ADC (IgG-MMAE), 100 nmol/L (IC90) Live/Dead Viability Dye (Thermo Fisher Scientific) and fluorescent
MMAE, vincristine, or paclitaxel for 18 hours. Treated L540cy cells antibodies targeting murine CD45 (RRID:AB_2563542), CD11c
were collected, lysed in RIPA buffer (Cell Signaling Technology) and (RRID:AB_313779), CD11b (RRID:AB_312791), Ly-6G (RRID:
centrifuged at 14,000–16,000 rpm for 10 minutes and stored at 20 C. AB_10643269), Siglec-F (RRID:AB_2904295), and MHC II
Cell pellets were resuspended in 4X BOLT LDS sample buffer (Thermo (RRID:AB_493147; BioLegend). Alternatively, portions of each
Fisher Scientific) and lysed by heating at 95 C for 5–10 minutes to tumor were weighed and lysed in RIPA buffer (Cell Signaling
produce cell lysates. Sample lysates were run on a Bis-Tris 4%–12% Technology) for cytokine analysis using the MILLIPLEX MAP
gradient gel at 140 V for 1 hour 40 minutes in MOPS buffer. The Bis- Human Cytokine/Chemokine Magnetic Bead Panel (Sigma).
Tris gel was then transferred onto a nitrocellulose membrane using an
iBlot2. Membranes were washed once in 1X TBS and incubated In vivo autologous lymphoblastoid cell line (LCL) PBMC
overnight at 4 C with antibodies against total and pIRE1, total and tumor model
phosphorylated c-Jun N-terminal kinase (pJNK), and activating tran- A total of 2.5106 Epstein-Barr virus (EBV)-transformed CD30þ
scription factor 4 (ATF4) (Cell Signaling Technology and Novus; LCLs, derived from a healthy donor, were implanted subcutaneously
RRID:AB_10145203, RRID:AB_2058748). Equal loading was con- into groups of eight nonobese diabetic (NOD)/SCID/gamma-chain
firmed with total actin or tubulin assessment (Cell Signaling Tech- mice (NSG) mice (RRID:IMSR_JAX:005557). For assessing PBMC-

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


nology, RRID:AB_330288, RRID:AB_2210548). Membranes were mediated rejection of LCL tumors, 0, 2.5, 5, or 10 million thawed
washed four times in 1X TBS-T for 5–10 minutes each. Signals were autologous donor PBMCs were given to mice intravenously 5 days
normalized against actin. after tumor implant following which tumor volumes were measured
periodically.
C/EBP homologous protein (CHOP) luciferase assay For experiments testing the effect of brentuximab vedotin on
Induction of CHOP was measured using a reporter system for responding immune cells, mice received a single subtherapeutic dose
CHOP activity according to the manufacturer’s instructions (Bright- of brentuximab vedotin or a non-binding ADC (human IgG-MMAE;
Glo Luciferase Assay System, Promega). In brief, 100,000 Mia-PaCa-2 1 mg/kg, i.p.) when tumor volumes averaged 250 mm3, then received
cells/well (Signosis; SL-0025-FP) were plated in a 96-well, flat-bottom 2.0106 autologous PBMCs 3 days post-dose. Four days following the
clear plate (aliquot 150 mL/well). At 24, 48, and 72 hours, plates were transfer of PBMCs, tumors were harvested from six mice. Tumor
removed from the incubator and allowed to come to room temper- RNA was purified using the RNeasy Plus Kit (Qiagen, Germany),
ature. 100 mL of media were removed from the wells and 100 mL of and RNA evaluated using the NanoString nCounter Human Immu-
Bright-Glo reagent was added to each well. Plates were shaken for at nology panel (NanoString Technologies). Data were processed and
least 2 minutes before measurement on an Envision plate reader analyzed using nSolver software (NanoString Technologies). Eleven
(PerkinElmer). days following transfer of PBMCs, tumors were harvested from
five mice, weighed, and manually dissociated through a 70-mm cell
Immune cell activation strainer for FACS analysis. Tumor cell suspensions were stained
Cancer cell/peripheral blood mononuclear cell (PBMC) co-cultures: with Zombie Aqua Viability Dye (BioLegend) followed by incubation
induction of innate and secondary T-cell responses following with fluorescently labeled antibodies targeting human CD19 (RRID:
co-culture of human PBMCs with cancer cells induced to undergo AB_2564143), CD2 (RRID:AB_2572066), CD8 (RRID:AB_2562790),
ICD following exposure to brentuximab vedotin or MMAE CD4 (RRID:AB_2572097), CD56 (RRID:AB_2563915), CD45
L540cy cancer cells were plated in 12-well plates and treated with (RRID:AB_2562057), programmed death-ligand 1 (PD-L1; RRID:
1 mg/mL of brentuximab vedotin, IgG-MMAE, or 0.1 mmol/L of AB_2563852), PD-1 (RRID:AB_2922606), and murine CD45.1
MMAE for 16 hours. Cells were harvested by moving samples to a (RRID:AB_2562565; BioLegend) in staining buffer (PBS, 2% FCS,
15-mL conical tube and centrifuging for 5 minutes at 400  g. Tissue 1% normal rabbit serum, 0.05% NaN3) at 4 C for 30 minutes. Cells
culture supernatants were removed and cells resuspended in com- were washed and resuspended in 120 mL of staining buffer for plate-
plete RPMI (10% FCS, 1 sodium pyruvate, 1 MEM NEAA, based flow cytometry using an Attune NXT flow cytometer. Total
1 GlutaMax; Gibco) at a total of 1107 cells/mL and plated at events were collected from 80 mL of sample, and flow cytometry-
250,000 cells/well in a 24-well plate. Human PBMC were thawed and measured cell concentrations were used to calculate numbers of
washed three times in complete RPMI and resuspended at a total of infiltrating immune cells. CD8þ T cells were identified as viability
1107 cells/mL. PBMCs were mixed with treated tumor cells 1:1 at dye negative, hCD45þ, mCD45.1, CD2þ, and CD8þ cells. Natural
500,000 cells/well and incubated at 37 C, 5% CO2, for 24 or 48 hours. killer (NK) cells were identified as viability dye negative, hCD45þ,
Tissue culture supernatants were harvested for analysis of innate and mCD45.1, CD2þ, and CD56þ cells. Experiments combining bren-
adaptive cell activation using the MILLIPLEX MAP Human Cyto- tuximab vedotin with nivolumab treatments were conducted as
kine/Chemokine Magnetic Bead Panel (Sigma). described above, with mice receiving two doses of nivolumab
(10 mg/kg, i.p.) 2 and 7 days after adoptive transfer of PBMCs.
L540cy xenograft
Five million L-540cy cells were implanted subcutaneously in Immunohistochemistry
25% Matrigel (Corning) in female (RRID:IMSR_TAC:CB17SC) severe The detection of infiltrating cells in the A20 syngeneic model was
combined immune deficient (SCID) mice (Taconic). When tumors performed as follows: mouse tissues were dissected and embedded in
reached 200 mm3, mice (n ¼ 5) were randomized into treatment OCT (TissueTek; #62550–01) and frozen in isopentane cooled in
groups and treated intraperitoneally (i.p.) with 3 mg/kg brentuximab liquid nitrogen. Sections were cut at 4 mm and stored frozen at 80 C.
vedotin, IgG-MMAE, or cAC10 antibody. Seventy-two hours after Slides with frozen tissue sections were thawed, fixed in acetone for

70 Mol Cancer Ther; 23(1) January 2024 MOLECULAR CANCER THERAPEUTICS


Brentuximab Vedotin and Immunogenic Cell Death

A Surface calreticulin B Surface calreticulin+/PI–

100 Untreated
Oxaliplatin
80 Etoposide
BV

Positive (%)
Untreated cAC10
60
Oxaliplatin
40
Etoposide

cAC10 20

BV
0
100 101 102 103 104 6 24 48
Time (h)

C ATP secretion D HMGB1 release

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


ns
*
* ns
* ns
8 *** * 4
* *
6 3
Fold change

Fold change

4 2

2 1

0 0
NT BV lgG-MMAE MMAE Etop Oxal NT BV lgG-MMAE MMAE Etop Oxal

Figure 1.
Brentuximab vedotin–treated Hodgkin lymphoma cells express ICD hallmarks. A, MFI histograms of surface CRT staining on live L540cy cells measured by flow
cytometry following treatment for 18 hours with the IC50 or IC90 concentrations of BV, oxaliplatin, etoposide, and cAC10. B, L540cy cell-surface expression of
CRT measured by flow cytometry following treatment over a 48-hour time course with BV, cAC10, oxaliplatin, or etoposide. C, Extracellular ATP measured from
L540cy cell-free supernatant following treatment for 18 hours with BV, IgG-MMAE, MMAE, oxaliplatin, or etoposide. D, Extracellular HMGB1 measured from L540cy
cell-free supernatant following treatment for 18 hours with BV, IgG-MMAE, MMAE, etoposide, or oxaliplatin. Representative data from three independent experiments.
Symbols represent biological replicates.  P < 0.05;   P < 0.001 (one-way ANOVA followed by Dunnett’s multiple comparisons test) compared with BV. Bars at the
mean. ANOVA, analysis of variance; ATP, adenosine trisphosphate; BV, brentuximab vedotin; cAC10, chimeric anti-CD30 antibody; CRT, calreticulin; Dox, doxorubicin;
Etop, etoposide; h, hour; HMGB1, non-histone chromatin-binding protein high-mobility group box 1; IC, inhibitory concentration; ICD, immunogenic cell death; IgG,
immunoglobulin G; MFI, mean fluorescence intensity; MMAE, monomethyl auristatin E; PI, propidium iodide; ns, not significant; NT, not treated.

10 minutes at 20 C, and then air-dried for 10 minutes. Immunos- (Biocare). Sections were quenched with 3% H2O2 and blocked with
taining was performed on the Bond-Max and BondIII autostainers 0.5% Casein and 5% Goat Serum in TBS. Mouse anti-human CD8
(Leica Biosystems, Inc.) using Bond Polymer Refine Detection (DAB) (Abcam RRID:AB_443686) was applied for 1 hour, washed, and
Kits (Leica; #DS9800). Endogenous peroxidase activity was blocked detected with Biotinylated Goat anti-Mouse IgG (The Jackson
using PeroxAbolish (BioCare Medical; #PXA969). Protein Block Laboratory, RRID:AB_2338569). Sections were then developed with
(Dako; #X0909) was used to block non-specific staining. Sections Vectastain Elite ABC HRP Kit (#PK-6100) and Vector ImmPact
were then incubated with Biotinylated Rat anti-Mouse CD8 clone DAB (#SK-4103).
4SM15 (eBioscience, RRID:AB_2572771) or Biotinylated Rat IgG2a
clone eBR2a (eBioscience, RRID:AB_470084) as isotope control at A20hCD30 cell line vaccination experiments
3 mg/mL. Bound antibody was detected using the Vectastain Elite ABC To generate human CD30-expressing A20 mouse lymphoma cells,
HRP Kit (Vector Laboratories) and developed with DAB, from the cells were transfected with plasmid constructs encoding human
Bond Polymer Refine Staining Kit (Leica). TNFRSF8 (NM_001243.4) and sgRNA/Cas9 for control under the
Formalin-fixed, paraffin-embedded LCL tumor tissues were sec- endogenous Rosa26 site for constitutive expression. FACS yielded a
tioned onto adhesive slides, deparaffinized, and rehydrated to dis- clonal population of A20 cells that stably expressed human CD30,
tilled water. Slides were steamed for antigen retrieval in Diva solution called A20hCD30.

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 71


Heiser et al.

lgG-
A B NT BV MMAE MMAE

hlgG- pIRE1
MMAE
IRE1

pJNK
MMAE

JNK

ATF4
BV
Actin
Nuclei Tubulin ER Merged L540cy

C D
NT BV
NT

pIRE

MMAE IRE
L540cy
Nuclei ER pIRE1 Merged

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


E L540cy XBP1 HDLM-2 XBP1 KARPAS 299 XBP1 A20hCD30 XBP1
30 50 **** ns 80 100 ***
**** **** ***
Positive (%)
Positive (%)

80

Positive (%)
40

Positive (%)
*** ns ** 60 *
20 **** **
30 60
40
10 20 40
10 20 20
0 0 0 0
NT IgG- BV MMAE NT IgG- BV MMAE NT IgG- BV MMAE NT IgG- BV MMAE
MMAE MMAE MMAE MMAE

L540cy ATF6 HDLM-2 ATF6 KARPAS 299 ATF6 A20hCD30 ATF6


50 **** 50 80 100 ***
*** ***
****
Positive (%)

**** *

Positive (%)
Positive (%)

40 40 80
Positive (%)

** 60 **** *
30 **** ns 60
30
40
20 20 40
10 10 20 20
0 0 0 0
NT IgG- BV MMAE NT IgG- BV MMAE NT IgG- BV MMAE NT IgG- BV MMAE
MMAE MMAE MMAE MMAE

L540cy pPERK HDLM-2 pPERK KARPAS 299 pPERK A20hCD30 pPERK


20 * 60 80 ****
ns ns * 30 ****
Positive (%)
Positive (%)

ns ****
Positive (%)

ns 60
**** ns
Positive (%)

15
40 ***
10 20 40
5 20 10 20
0 0 0 0
NT IgG- BV MMAE NT IgG- BV MMAE NT IgG- BV MMAE NT IgG- BV MMAE
MMAE MMAE MMAE MMAE

F NT MMAE Paclitaxel Vincristine NT MMAE Paclitaxel Vincristine

pIRE1 pJNK

IRE1 JNK

G Dose-CHOP-luciferase induction H CHOP-luciferase activity


4 15
MMAE NT
Paclitaxel MMAE
Fold over pre-dose

3 Vincristine Paclitaxel
Fold change

10 Vincristine

2
5
1

0 0
0.1 1 10 100 1,000 0 24 48 72 96
nmol/L Hours post-dose

72 Mol Cancer Ther; 23(1) January 2024 MOLECULAR CANCER THERAPEUTICS


Brentuximab Vedotin and Immunogenic Cell Death

A20hCD30 cells were cultured in RPMI-1640 with 10% FBS, ciences). Cells were then washed with FACS buffer þ Annexin V
10 mmol/L HEPES, 1 mmol/L sodium pyruvate, penicillin (100 U/mL), binding buffer before fixation and permeabilization using the True
and streptomycin (100 mg/mL). A20hCD30 cells were treated with Nuclear Kit, 96-well plate protocol (BioLegend). Overnight intracel-
1 mg/mL brentuximab vedotin or 100 nmol/L mc–vc–MMAE for lular staining at 4 C was then performed upon the addition of 100 mL
4 days. To prepare dying cells for immunization, treated A20hCD30 of permeabilization buffer containing antibodies to each well. A list of
cells were overlaid atop Histopaque and centrifuged at 2,000  g for antibodies with corresponding RRIDs used for intracellular staining is
30 minutes. Dead and dying cells were pelleted underneath the provided in Supplementary Table S1. Cells were then washed three
Histopaque layer and viability was assessed to be <20% live cells by times in permeabilization buffer, resuspended in PBS, and analyzed on
trypan blue exclusion. Dead and dying A20hCD30 cells were resus- an Attune Flow Cytometer.
pended in PBS and a total of 2  106 cells were injected into the
peritoneum of immune-competent BALB/c mice (n ¼ 8). Flash-frozen Statistical analysis
A20hCD30 cells were prepared by submerging cells in liquid nitrogen for Descriptive statistics were used throughout. Where applicable,
10 seconds, followed by immersion in 37 C water until completely significance was determined by an ANOVA with Sidak or Dunnett’s
thawed. The liquid nitrogen freeze-thaw process was repeated five multiple comparisons tests, calculated using GraphPad Prism 8.1
times. Fourteen days later, mice received a second immunization with (RRID:SCR_000306).
dead and dying cells prepared in the same manner.
Twenty-one days after initial immunization, mice were subcuta- Data and materials availability
neously implanted with a total of 5  106 wild-type (WT) A20 cells and All available data are presented.

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


monitored for tumor growth. In some experiments, mice were treated
with four administrations of anti–PD-1 over 2 weeks (1 mg/kg;
BioLegend, RRID:AB_2783090). Results
Immune-competent BALB/c mice (n ¼ 4) were immunized with Brentuximab vedotin–treated Hodgkin lymphoma (HL) cells
brentuximab vedotin-killed or flash-frozen–killed A20hCD30 cells as express ICD hallmarks
previously described. Sixteen weeks after initial immunization, spleens Cell surface CRT expression, the first hallmark of ICD, was eval-
were harvested from immunized mice or naive BALB/c mice and uated on CD30þ L540cy cells to determine whether cell death induced
manually homogenized. Splenic homogenates were combined from by brentuximab vedotin is consistent with ICD induction. The L540cy
four mice per immunization group and CD3þ T cells were isolated cell line is a derivative of the CD30þ HL L540 line adapted for growth
using the EasySep Mouse T-cell Enrichment Kit (Stem Cell Technol- as a xenograft. L540cy cells were treated with brentuximab vedotin,
ogies). One million CD3þ T cells were administered intravenously into IgG-MMAE, oxaliplatin (a known ICD inducer; ref. 12), or etoposide
WT A20 tumor-bearing NSG mice when tumors averaged 100 mm3 (which does not induce ICD; ref. 27). After 18 hours of treatment, cells
(n ¼ 5–8). Tumors were subsequently monitored for growth and at were harvested, stained for cell-surface CRT expression, and levels
study takedown for CD8þ T-cell content by flow cytometry. were analyzed by flow cytometry. The mean fluorescence intensity of
surface CRT at 18 hours was increased in live cells (Annexin V/PI)
Intracellular staining of UPR markers by flow cytometry after brentuximab vedotin treatment, whereas IgG-MMAE or etopo-
Cells were harvested 48 hours following treatment with test articles. side-treated cells exhibited minimal changes in surface CRT (Fig. 1A).
Supernatants were transferred to round-bottom plates to collect non- Furthermore, the proportion of brentuximab vedotin–treated cells
adherent cells. Versene (200 mL; Thermo Fisher Scientific) was then expressing surface CRT was increased at 48 hours (Fig. 1B). Impor-
added to each well, and upon detachment, cells were transferred to the tantly, similar increases in expression of cell surface CRT were
corresponding round-bottom plates used to collect non-adherent cells. observed in three separate human lymphoma cell lines and the murine
Cells were pelleted by spinning at 1,500 rpm for 5 minutes. Plates were A20 tumor cell line engineered to express human CD30 (A20hCD30;
decanted, washed with PBS and, following an additional spin, cells Supplementary Fig. S2A), demonstrating a common effect across
were stained with Live/Dead Yellow cell viability dye (Thermo Fisher multiple target cell lines.
Scientific) per the manufacturer’s protocol. Surface staining for calre- The second hallmark signature of ICD induction is active secretion
ticulin and Annexin V was then performed in FACS buffer (2% of ATP through pannexin channels. Release of ATP serves as a potent
FBS/PBS) with the addition of Annexin V binding buffer (BD Bios- chemotactic signal recruiting immune cells to the inflamed site (52).

Figure 2.
Brentuximab vedotin–induced ICD is associated with severe ER stress. A, Fluorescent staining of tubulin, ER, and nuclei in L540cy cells treated for 16 hours with
human IgG-MMAE, MMAE, or BV. B, Western blot analysis of ER stress markers in L540cy cells treated for 18 hours with BV, IgG-MMAE, and MMAE. C, Spatial
relationship of ER stress markers with the ER network in L540cy cells treated for 18 hours with MMAE. D, Western blot analysis of pIRE1 and IRE1 expressions in L540cy
cells treated for 18 hours with a titration of BV. E, Intracellular detection of the proportion of viable lymphoma cells (L540cy, HDLM-2, KARPAS 299, and A20hCD30)
upregulating UPR-associated proteins XBP1, ATF6, and pPERK by flow cytometry 48 hours after treatment with IC20–IC50 doses of the indicated agents.
F, Representative western blot analysis of pIRE1, IRE1, pJNK, and JNK expression in L540cy cells treated for 18 hours with MMAE, vincristine, or paclitaxel. G, Dose–
CHOP luciferase induction in Mia-PaCa-2 cells treated with MMAE, vincristine, or paclitaxel in vitro. H, CHOP-luciferase activity in Mia-PaCa-2 cells treated
intratumorally with MMAE, vincristine, or paclitaxel in vivo. Western blot analysis band intensity quantification provided in Supplementary Fig. S3. Symbols
represent biological replicates.  P < 0.05;   P < 0.01;    P < 0.001;     P < 0.0001, (one-way ANOVA followed by Dunnett’s multiple comparisons test)
compared with BV (for human lines), or MMAE (for A20hCD30). Bars at the median. ANOVA, analysis of variance; ATF4, activating transcription factor 4; ATF6,
activating transcription factor 6, BV, brentuximab vedotin; CHOP, C/EBP homologous protein; ER, endoplasmic reticulum; hIgG, human immunoglobulin G; IC,
inhibitory concentration; IgG, immunoglobulin G; IRE1, inositol-requiring enzyme 1; JNK, c-Jun N-terminal kinase; MMAE, monomethyl auristatin E; ns, not
significant; NT, not treated; pIRE1, phosphorylated IRE1; pJNK, phosphorylated JNK; pPERK, phosphorylated protein kinase RNA-activated–like endoplasmic
reticulum kinase; UPR, unfolded protein response; XBP1, X-box binding protein 1.

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 73


Heiser et al.

A Human dendritic cells Human T cells


MIP-1α IP10/CXCL10 IFNγ TNFα
* ns ns
ns ns ns
ns
** *
* *
ns ns
3,000 1,200 1,500 * 500
ns
ns 400
900
2,000 1,000
pg/mL

pg/mL

pg/mL

pg/mL
300
600
200
1,000 500
300
100

0 0 0 0

Ig M BV
-M AE

O E
Et l
op
Ig M B V
-M AE

O E
Et l
op

Ig M BV
-M AE

O E
Et l
op

Ig M BV
-M AE

O E
Et l
op
xa
xa

xa

xa
A
A

A
G M
M
G M
M

G M
M

G M
M

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


B Dendritic cells Dendritic cells/tumor Neutrophils/immune cells
ns
***
Percentage of CD11c/total cells
Percentage of CD11c of CD45+

*** ns

Percentage of Ly6g/CD45+
25 5 15
***
20 4
*
10
15 3

10 2
5
5 1

0 0 0
cAC10 IgG-MMAE BV cAC10 IgG-MMAE BV cAC10 IgG-MMAE BV

Intratumoral IP10/CXCL10 Intratumoral MIP-1α Intratumoral TNFα


C
**
* ns *
5,000 1,000 250 ns
ns
4,000 800 200
pg/mL

pg/mL

pg/mL

3,000 600 150

2,000 400 100

1,000 200 50

0 0 0
cAC10 IgG-MMAE BV cAC10 IgG-MMAE BV cAC10 IgG-MMAE BV

D Serum IP10/CXCL10 Serum MIP-1α Serum TNFα


ns ns
ns
600 * 80 ns 15 *
60
400 10
pg/mL

pg/mL

pg/mL

40
200 5
20

0 0 0
cAC10 IgG-MMAE BV cAC10 IgG-MMAE BV cAC10 IgG-MMAE BV

74 Mol Cancer Ther; 23(1) January 2024 MOLECULAR CANCER THERAPEUTICS


Brentuximab Vedotin and Immunogenic Cell Death

L540cy cells were treated with brentuximab vedotin, a non-binding and pJNK were minimally expressed with controls (no treatment and
ADC, free MMAE, oxaliplatin, or etoposide for 18 hours. Cell-free IgG-MMAE; Fig. 2B and D). Furthermore, microscopy showed pIRE1
supernatant was collected and analyzed for ATP levels that were to be spatially localized with the structurally disrupted ER in MMAE-
normalized to levels from untreated cells. Following treatment with treated L540cy cells compared with control, corroborating a potent
brentuximab vedotin or free MMAE, extracellular ATP levels approx- UPR response (Fig. 2C). Further evidence for activation of the three
imated or were greater than those obtained following treatment arms of the UPR was provided by staining of intracellular XBP1, ATF6,
with oxaliplatin (Fig. 1C). Treatment with etoposide, a cytotoxic and phosphorylated protein kinase RNA-activated-like ER kinase by
agent that does not induce ICD, did not induce robust ATP secretion flow cytometry for CD30-expressing lymphoma lines 48 hours after
from dying L504cy cells. Heightened ATP secretion following bren- treatment with brentuximab vedotin or MMAE (Fig. 2E). Significant
tuximab vedotin and MMAE treatment was also observed with increases in UPR stress markers were observed for each cell line treated
additional lymphoma lines KARPAS 299 and the murine A20hCD30 with brentuximab vedotin compared with untreated controls. Com-
(Supplementary Fig. S2A). pared with human lymphoma lines, the engineered A20hCD30 synge-
Finally, extracellular HMGB1 elicits inflammatory responses as a neic lymphoma expressed much lower levels of CD30 (Supplementary
ligand for TLR4, TLR2, and advanced glycosylation end product- Fig. S2B) and showed stronger UPR induction from free MMAE
specific receptor (AGER, RAGE), triggering potent NF-kB responses treatment compared with brentuximab vedotin.
in immune cells (1, 6, 10). After 18 hours of treatment with brentux- The effects of using microtubule-destabilizing agents (MMAE
imab vedotin, free MMAE, etoposide, or oxaliplatin, HMGB1 was and vincristine) versus a stabilizing agent (paclitaxel) were inves-
elevated in the cell-free supernatant of L540cy cells (Fig. 1D). tigated to determine whether the underlying mechanism behind ER

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


Increased release of HMGB1 was also observed for KARPAS 299, stress induction was associated with collapse of the microtubule
HDLM-2, and A20hCD30 cell lines following brentuximab vedotin or network. Although IRE1 phosphorylation occurred after treatment
MMAE treatment at 72 hours (Supplementary Fig. S2A). Elevation of with all agents, indicating acute ER stress and initial activation
HMGB1, a late-stage marker of ICD, was seen upon etoposide of stress responses, only the microtubule-destabilizing agents
treatment, but occurred in the absence of robust CRT exposure and MMAE and vincristine elicited downstream JNK phosphorylation
ATP release, indicating that molecules like MMAE are unique in their (Fig. 2F). Pixel densities for all western blot analysis images are
ability to drive all three markers of ICD. Overall, these results provided in Supplementary Fig. S3A–S3D. Robust pIRE1 and pJNK
demonstrate that treatment of CD30þ lymphoma cell lines with upregulation was also observed following treatment with brentux-
brentuximab vedotin or MMAE was associated with the presentation imab vedotin or MMAE in additional lymphoma lines by intracel-
of three hallmarks of ICD. lular flow cytometry (Supplementary Fig. S4A). Sustained activation
of the UPR response ultimately leads to the initiation of ICD
MMAE-induced microtubule disruption by brentuximab vedotin through the induction of CHOP. To assess CHOP induction from
leads to severe ER stress microtubule-stabilizing versus -destabilizing compounds, an ER
The ER is a dynamic cellular organelle that expands and contracts stress reporter system of Mia-PaCa-2 cells transduced with a
depending on the functional needs of the cell (53, 54). Expansion and CHOP-driven luciferase was employed. In this system, paclitaxel-
contraction occur along the tubulin highway (55) and catastrophic driven microtubule stabilization did not drive equivalent CHOP
disruption of that network can inhibit normal ER function (56). As induction as the microtubule-destabilizing compounds MMAE and
such, the relationship between MMAE-mediated disruption of the vincristine, despite visible cell death (Fig. 2G). CHOP induction
microtubule network and the resultant effects on ER structure and was confirmed in three additional lymphoma lines treated with
stress response in treated cells was investigated. Compared with brentuximab vedotin or free MMAE by intracellular flow cytometry
control (IgG-MMAE), treatment of L540cy cells with brentuximab (Supplementary Fig. S4B).
vedotin or free MMAE led to a striking qualitative disorganization of Induction of ER stress with microtubule-disrupting agents, includ-
the perinuclear ER network (Fig. 2A). Disruption of protein folding in ing MMAE, was validated in vivo by engrafting CHOP-luciferase Mia-
the ER results in a well-defined stress response, referred to as the UPR, PaCa-2 cells into NSG. Xenografts were treated intratumorally with
purposed for re-establishing ER homeostasis. To determine whether MMAE, vincristine, or paclitaxel and monitored daily for luciferase
the observed disorganization of the ER network occurs concomitant activity. Soon after administration, CHOP induction, as monitored by
with the UPR response, L540cy cells were treated for 18 hours with luciferase activity, was observed from tumors treated with MMAE or
brentuximab vedotin or free MMAE. After 18 hours, there was a rapid vincristine, peaking at 72 to 96 hours after treatment (Fig. 2H).
induction and activation of multiple arms of the UPR, including Notably, luciferase was detectable 24 hours after treatment, indicating
increased pIRE1, downstream pJNK, and ATF4 expression; pIRE1 that the response happened quickly and increased over time.

Figure 3.
Proinflammatory chemokine and cytokine production in human dendritic and T cells and BV-induced ICD in L540cy xenografts. A, Chemokine and cytokine
measurements by immunoassay (Luminex) following washing out of treatment agents. B, Recruitment of CD11cþ DCs compared with Ly6gþ cells into a L540cy
xenograft 72 hours after administration of a single dose of BV, IgG-MMAE, or cAC10 (murine model; n ¼ 5); levels of immune cells were determined by flow cytometry.
C, Intratumoral murine cytokine activity in mice harboring L540cy xenografts 72 hours after administration of a single dose of BV, IgG-MMAE, or cAC10 (n ¼ 5);
cytokines were measured by immunoassay (Luminex). D, Upregulation of murine serum chemokines and cytokines in mice harboring L540cy xenografts 72 hours
after administration of a single dose of BV, IgG-MMAE, or cAC10 (n ¼ 5); cytokines were measured by immunoassay (Luminex). Representative data from two
independent experiments, each symbol represents individual mice (n ¼ 4–5 per group).  P < 0.05;   P < 0.01;   P < 0.001, (one-way ANOVA followed by Dunnett’s
multiple comparisons test) compared with BV treatment. Bars represent the mean and SE. ANOVA, analysis of variance; BV, brentuximab vedotin; cAC10, chimeric
anti-CD30 antibody; CCL2, chemokine (C-C motif) ligand 2; CXCL10, C-X-C motif chemokine ligand 10; Etop, etoposide; ICD, immunogenic cell death; IFN, interferon;
IgG, immunoglobulin G; IP10, interferon gamma-induced protein 10; MIP-1a, macrophage inflammatory protein-1a; MMAE, monomethyl auristatin E; ns, not
significant; NT, not treated; Oxal, oxaliplatin; SE, standard error; TNF, tumor necrosis factor.

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 75


Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024

MOLECULAR CANCER THERAPEUTICS


76 Mol Cancer Ther; 23(1) January 2024
Heiser et al.
Brentuximab Vedotin and Immunogenic Cell Death

Treatment with microtubule-stabilizing paclitaxel did not elicit lucif- significant or trending increases in intratumoral and serum murine
erase activity, but inhibited tumor growth, indicating that antitumor proinflammatory cytokines TNFa, IP10, and MIP-1a (Fig. 3C
activity of paclitaxel did not rely on ER stress induction. and D). Furthermore, tumor-derived cytokine concentrations were
notably higher than those detected in circulation, consistent with
Immune cell activation by cells undergoing ICD in response to tumor-localized production. In vitro activation of human immune
brentuximab vedotin cells and recruitment of murine DCs in the L540Cy xenograft
After confirming that cells treated with brentuximab vedotin exhibit model following treatment with brentuximab vedotin are consis-
signs of severe ER stress and express the key ICD hallmarks, we tent with ICD induction and highlight the potential for driving
questioned whether the dying cells were able to activate innate and focused immune activation in the TME following treatment with
adaptive immune responses. To this end, brentuximab vedotin–killed MMAE-based ADCs.
L540cy cells were fed to human monocyte-derived CD11cþ dendritic
cells (DC) in vitro and proinflammatory chemokine and cytokine Brentuximab vedotin–induced ICD promotes CD8þ T-cell and
production were measured in supernatants. Notably, macrophage NK-cell recruitment to human LCL tumors
inflammatory protein-1a (MIP-1a) and IFN gamma–induced protein The ability of brentuximab vedotin treatment to promote proin-
10/C-X-C motif chemokine ligand 10 (IP10/CXCL10) were increased flammatory cytokine production in xenografts is consistent with ICD-
in supernatants from DCs that were fed cells treated with brentuximab driven activation of innate immunity. To further investigate the impact
vedotin, MMAE, etoposide, or oxaliplatin compared with IgG-MMAE of this inflammatory response on the recruitment and activation of
(Fig. 3A). Treatment with the non-targeting ADC did not significantly antigen-specific cytotoxic T cells to the TME, we used a humanized
LCL/PBMC model. This model pairs EBV-transformed CD30þ LCL

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


promote proinflammatory mediators, showing that delivery of MMAE
to tumor cells was required for driving proinflammatory cytokines tumors with adoptively transferred EBV-reactive autologous PBMCs to
from human DCs. Oxaliplatin and etoposide, which were both shown evaluate antigen-specific T-cell responses in vivo. In this model, intra-
to induce HMGB1 release from L540Cy cells (Fig. 1D), also drove venous transfer of autologous PBMCs results in a titratable, immune-
proinflammatory cytokine secretion consistent with this component of mediated elimination of the tumor as measured by tumor volume
ICD. In addition, when brentuximab vedotin- and MMAE-treated reduction (Fig. 4A). To evaluate whether treatment of LCL tumors
cells were co-cultured with DCs and autologous T cells, increased with brentuximab vedotin can influence the innate and adaptive arms of
production of T-cell cytokines IFNg and TNFa was observed, com- the immune system, mice with established LCL tumors (200 mm3) were
pared with IgG-MMAE and a trending increase compared with given a subtherapeutic dose of brentuximab vedotin (the highest dose
etoposide-treated cells (Fig. 3A). Together, the increase in DC- and without consistent tumor reduction; 1 mg/kg) followed by adoptive
T-cell–derived proinflammatory cytokines demonstrates the ability of transfer of autologous PBMCs 3 days later. Tumors from brentuximab
MMAE- and brentuximab vedotin–killed tumor cells to promote APC vedotin–treated and control groups were harvested for analysis 4 and
activity. 11 days after PBMC transfer (Fig. 4B). NanoString analysis of brentux-
The host innate immune response to brentuximab vedotin– imab vedotin-treated LCL tumors harvested 4 days after PBMC transfer
treated tumors was investigated in vivo in a human L540cy xeno- showed increased lysosome-associated membrane glycoprotein 3 tran-
graft model in SCID mice. SCID mice lack T and B cells but retain script consistent with UPR-induced autophagy in tumor cells (Fig. 4C;
innate immune cells enabling evaluation of APC activation and ref. 57). UPR-induced autophagy in tumor cells undergoing ICD drives
recruitment following ADC killing of xenograft tumors. Although the hallmark release of extracellular ATP that signals via purinergic
brentuximab vedotin treatment did not significantly influence the receptors to activate the NLRP3 inflammasome in neighboring macro-
scale of immune infiltration into the tumor by 72 hours, it did begin phages (22, 58, 59). Consistent with increased extracellular ATP, LCL
to reshape the immune landscape as shown by increased propor- tumors treated with brentuximab vedotin showed increased NLRP3
tions of antigen-presenting CD11cþ DCs in tumors relative to transcript (Fig. 4C). Finally, signatures reflecting early innate (CD11b,
Ly6gþ neutrophils (Fig. 3B). Skewing of the available innate CXCL9) and cytotoxic immune cell (IFNg, Granzyme B) activity and
immune compartment toward mature DCs (CD11cþ DCs) may recruitment were strongly upregulated in brentuximab vedotin-treated
reflect recruitment of DCs into the tumor or differentiation of samples compared with non-targeted ADC and antibody alone controls
infiltrating monocytes following tumor cell killing by brentuximab (Fig. 4C). Eleven days after PBMC transfer, flow cytometry of processed
vedotin. Furthermore, consistent with innate immune cell activa- tumors showed significantly increased CD8þ T cells and CD56þ NK
tion and recruitment following ICD induction, brentuximab vedo- cells in brentuximab vedotin–treated mice, demonstrating enhanced
tin treatment, compared with IgG-MMAE and cAC10, led to recruitment, and/or proliferation of innate and adaptive cytotoxic

Figure 4.
Brentuximab vedotin treatment of humanized mice bearing LCL tumors enhances intratumoral immune activation. NSG mice (n ¼ 5) harboring CD30þ LCL tumors
were injected intravenously with autologous donor PBMCs 5 days after tumor implantation. A, Tumor volume was measured over time in NSG mice (n ¼ 5 per group)
receiving no PBMCs and 2.5, 5, and 10 million PBMCs. B, Mice were treated with BV, cAC10, or a non-binding control (human IgG-MMAE) 3 days before receiving a total
of 2.0  106 PBMCs (n ¼ 10 per group). Tumors were collected for analysis of immune activity 4 and 11 days after transfer of PBMCs (n ¼ 5 per time-point). C, Immune-
related transcripts (NanoString) were measured 4 days following PBMC transfer in NSG mice treated with BV, cAC10, or a non-binding control (IgG-MMAE) 3 days
before PBMC transfer. D, Tumors taken 11 days after PBMC transfer in NSG mice treated with BV or a non-binding control (IgG-MMAE) 3 days before PBMC transfer
were processed and evaluated by flow cytometry for immune cell infiltration. Each symbol represents NanoString analysis or flow cytometry of immune cells from
individual mice (n ¼ 4–7 per group).  P < 0.05;   P < 0.01;    P < 0.001;     P < 0.0001 (one-way ANOVA followed by Dunnett’s multiple comparisons test)
compared with BV treatment. Bars represent the mean and SE. ANOVA, analysis of variance; BV, brentuximab vedotin; cAC10, chimeric anti-CD30 antibody; CXCL9,
C-X-C motif chemokine ligand 9; GZMB, granzyme B; IFNg, interferon gamma; hIgG, human immunoglobulin G; IgG, immunoglobin G; ITGAM, integrin alpha M;
LAMP3, lysosomal-associated membrane glycoprotein 3; LCL, lymphoblastoid cell line; MMAE, monomethyl auristatin E; NK, natural killer, NLRP3, nucleotide-binding
domain, leucine-rich–containing family pyrin domain–containing-3; NOD, nonobese diabetic; NSG, NOD SCID gamma; PBMC, peripheral blood mononuclear cells;
SCID, severe combined immune deficient; SE, standard error; TIL, tumor-infiltrating lymphocyte; TNF, tumor necrosis factor.

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 77


Heiser et al.

A 1,250
Untreated
1,250
MMAE-killed A20
1,250
BV-killed A20
1,250
Freeze-thawed A20

Tumor volume (mm3)


Tumor volume (mm3)

Tumor volume (mm3)

Tumor volume (mm3)


1,000 1,000 1,000 1,000
1/5 CR 6/8 CR 5/8 CR 3/8 CR
750 750 750 750
500 500 500 500
250 250 250 250
0 0 0 0
0 7 14 21 28 35 42 0 7 14 21 28 35 42 0 7 14 21 28 35 42 0 7 14 21 28 35 42
Days post-tumor implant Days post-tumor implant Days post-tumor implant Days post-tumor implant

B Non-immunized
Immunized with
MMAE-killed A20 cells
Immunized with
BV-killed A20 cells

200 µm

C Untreated MMAE-killed A20 BV-killed A20

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


Tumor volume (mm3)

Tumor volume (mm3)

Tumor volume (mm3)


1,000 1,000 1,000
0/9 CR 5/9 CR 4/9 CR
750 750 750
500 500 500
250 250 250
0 0 0
0 7 14 21 28 35 0 7 14 21 28 35 0 7 14 21 28 35
Days post-tumor implant Days post-tumor implant Days post-tumor implant

Anti–PD-1 MMAE-killed A20 + anti–PD-1 BV-killed A20 + anti–PD-1


Tumor volume (mm3)

Tumor volume (mm3)

Tumor volume (mm3)


1,000 1,000 1,000
750 2/9 CR 750 9/9 CR 750 8/9 CR

500 500 500


250 250 250
0 0 0
0 7 14 21 28 35 0 7 14 21 28 35 0 7 14 21 28 35
Days post-tumor implant Days post-tumor implant Days post-tumor implant

D No T Cells T cells from naïve BALB/c T cells from BV-killed A20 T cells from freeze-thawed A20
Tumor volume (mm3)

Tumor volume (mm3)

Tumor volume (mm3)

Tumor volume (mm3)

1,250 1,250 1,250 1,250


1,000 1,000 1,000 1,000
750 750 750 750
500 500 500 500
250 250 250 250
0 0 0 0
0 5 10 15 20 0 5 10 15 20 0 5 10 15 20 0 5 10 15 20
Days post-tumor implant Days post-tumor implant Days post-tumor implant Days post-tumor implant

Tumor AUC from


E T-cell adoptive transfer F Intratumoral CD8
ns
***
ns
15,000 8
total tumor cells (CD45+)

**
AUC to day 19

Percentage of

* 6

10,000 4

2
5,000
0
aw e-
ve

SG
aw e-

ve
Fr d

ed
ed

BV
ille

th ez
th z


ee

N
N

e
-k

N
Fr
BV

78 Mol Cancer Ther; 23(1) January 2024 MOLECULAR CANCER THERAPEUTICS


Brentuximab Vedotin and Immunogenic Cell Death

cells (Fig. 4D; Supplementary Fig. S5). Results from the LCL/PBMC To further demonstrate the impact of MMAE-induced ICD on the
xenograft model are consistent with upregulation of inflammatory long-lived cytotoxic T-cell response, CD3þ T cells were isolated from
murine chemokines in L540cy xenografts treated with brentuximab the spleens of tumor cell-immunized BALB/c mice 5 months after
vedotin (Fig. 3) and substantiate enhancement of both innate cell and vaccination and adoptively transferred to NSG mice harboring existing
antigen-specific T-cell responses following brentuximab vedotin–driven A20 tumors (approximately 100 mm3). Use of NSG mice precludes
ICD induction. host antitumor immunity, enabling the assessment of adoptively
transferred T-cell activity. NSG mice receiving no T cells, T cells
Brentuximab vedotin–mediated ICD drives T-cell memory and transferred from naive mice, or T cells from mice immunized with
protects mice from subsequent tumor challenge control freeze-thawed–killed tumor cells showed minimal tumor
The “gold-standard” experiment for assessing a compound’s ability control and were sacrificed at study take down (day 19), when all
to elicit functional ICD involves vaccination of mice with treated control tumors exceeded 1,000 mm3. In contrast, mice receiving T cells
syngeneic tumor cells followed later by tumor rechallenge. Bona fide from mice immunized with brentuximab vedotin–killed A20hCD30
ICD-inducing compounds drive lasting antigen-specific memory tumor cells showed significantly slowed tumor growth up to day 19 as
responses that prevent tumor growth upon subsequent implant with determined by the tumor volume area under the curve (Fig. 5D and E).
the same cell line (6, 12, 60). To validate ICD induction and tumor- Furthermore, mice receiving T cells from brentuximab vedotin–
specific adaptive immune responses following brentuximab vedotin immunized mice showed a trend toward increased CD8þ T-cell
treatment, the human CD30-engineered syngeneic A20 B-cell lym- infiltration in A20 tumors at take down (Fig. 5F). Together, these
phoma model (A20hCD30) was used. A20hCD30 cells were killed in vitro results are consistent with enhanced T-cell immunity elicited by

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


with brentuximab vedotin or MMAE and injected into WT BALB/c known ICD inducers in similar models alone and in combination
mice. As a negative control, A20hCD30 cells were flash-frozen in liquid with anti-PD-1 (21, 27, 39) and provide evidence that MMAE is an
nitrogen providing tumor-associated antigens in the absence of ICD-inducing agent capable of promoting long-lived T-cell responses.
ICD induction. Mice were immunized two times, and 21 days after
the initial immunization mice were implanted with WT A20 lympho- Combination of brentuximab vedotin treatment and PD-1
ma cells and monitored for tumor growth. The majority of mice inhibition improves antitumor immunity in the autologous
immunized with brentuximab vedotin- or MMAE-killed cells began LCL/PBMC xenograft model
to reject the tumors around 7 days after implant, resulting in 5/8 and The combination of brentuximab vedotin and PD-1 inhibition was
6/8 complete responses (CR)s, respectively, consistent with a also evaluated in the humanized autologous LCL/PBMC xenograft
primed adaptive immune response. In contrast, unimmunized naive model. In this model, LCL tumor clearance is mediated by autologous
mice and mice immunized with control freeze-thawed cells showed EBV-specific cytotoxic T-cell responses. As previously described,
accelerated tumor growth and fewer CRs, 1/5 and 3/8 CRs, respec- treatment of LCL tumors with a subtherapeutic dose of brentuximab
tively (Fig. 5A). Consistent with memory T-cell generation fol- vedotin augmented infiltration of cytotoxic T and NK cells and
lowing vaccination, mice that had previously received brentuximab increased production of IFNg (Fig. 4C and D). Inhibitory PD-L1
vedotin- or MMAE-killed cells showed rapid infiltration and expression can be driven though IFNg and the JAK/STAT pathway
increased murine CD8þ T cells within A20 tumors shortly after following activation of cytotoxic T cells and may limit antitumor
implant (Fig. 5B). immunity (62). Flow cytometric evaluation of on-study LCL tumor
Clinical response rates to PD-1/PD-L1 therapies correlate with the cells and intra-tumoral CD8þ T cells in this model showed high
presence of CD8þ T cells in the TME (61). Given that T cells from mice expression of PD-L1 and PD-1, respectively, supporting the presence
immunized with brentuximab vedotin–killed tumor cells could confer of this inhibitory axis in LCL tumors (Fig. 6A).
anamnestic antitumor protection, the combination with PD-1 inhi- PBMC-humanized mice receiving a subtherapeutic dose of bren-
bition was investigated. BALB/c mice were immunized with brentux- tuximab vedotin showed accelerated LCL tumor rejection relative to
imab vedotin- or MMAE-killed A20hCD30 cells, implanted with WT untreated humanized mice, consistent with enhanced T-cell activity
A20 tumor cells 14 days later, and then given anti–PD-1 7 days after (Fig. 6B and C). Although blocking of human PD-1 with nivolumab
tumor implantation. Although immunization with brentuximab vedo- did not significantly accelerate tumor rejection as a single agent, the
tin- or MMAE-killed A20 cells alone again provided antitumor combination of brentuximab vedotin and nivolumab showed superior
immunity, the addition of anti–PD-1 robustly increased the propor- tumor rejection compared with either treatment alone (Fig. 6B and C).
tion of CRs (Fig. 5C). These results demonstrate that MMAE-induced Results from the humanized LCL tumor model are consistent with the
ICD can be combined with PD-1 blockade to improve antitumor murine A20 vaccination model and further substantiate the comple-
immunity. mentarity of MMAE-driven ICD and PD-1 inhibition. In addition to

Figure 5.
Brentuximab vedotin–induced ICD protects against subsequent tumor challenges. A20 lymphoma cells expressing human CD30 (A20hCD30) were treated with BV or
MMAE in vitro, and dying cells were used to immunize wild-type BALB/c mice. Cells flash-frozen in liquid nitrogen were injected as a non-ICD control. A, Mice (n ¼ 8
per treatment group) immunized with BV-killed, MMAE-killed, or flash-frozen A20hCD30 cells were challenged with wild-type A20 cells on day 14 and monitored for
tumor growth. B, Detection of CD8þ T cells within the tumor by IHC after 9 days. C, BALB/c mice (n ¼ 9 per group) were immunized with BV- or MMAE-killed A20 cells
and rechallenged 14 days later. Anti–PD-1 (1 mg/kg) was administered on days 6, 9, 14, and 17 after tumor challenge and tumor growth was monitored. Numbers in
figures reflect surviving mice out of total at day 29. D, A20 tumor-bearing NSG mice (n ¼ 5–8 per treatment group) received CD3þ T cells from mice immunized with
BV-killed or flash-frozen–killed A20 cells, or T cells from unimmunized mice (naive) and monitored for tumor growth. E, Tumor volume AUC for mice receiving T cells
from mice immunized with brentuximab vedotin–killed A20hCD30 tumor cells. F, CD8þ T-cell infiltration. Representative data from two independent experiments,
each symbol represents an individual mouse.  P < 0.05;   P < 0.01;    P < 0.001, (one-way ANOVA followed by Dunnett’s multiple comparisons test) compared
with BV treatment. Bars represent the mean. ANOVA, analysis of variance; AUC, area under the curve; BV, brentuximab vedotin; CR, complete response; ICD,
immunogenic cell death; IHC, immunohistochemistry; MMAE, monomethyl auristatin E; NOD, nonobese diabetic; ns, not significant; NSG, NOD SCID gamma; NT, not
treated; PD-1, programmed cell death protein 1; SCID, severe combined immune deficiency.

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 79


Heiser et al.

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


Figure 6.
Protective immunity from brentuximab vedotin- or MMAE-mediated immunization is enhanced by anti–PD-1 treatment. A, Representative expression of PD-1 and
PD-L1 on T cells and LCL tumor cells, respectively, on day 35 by flow cytometry in the autologous PBMC/LCL tumor model. Staining demonstrates the presence
of an active PD-1/PD-L1 axis between T cells and LCL tumor cells. B, LCL tumor volumes were analyzed for treatment conditions relative to mice that received
only PBMCs. Tumor-bearing mice that received PBMCs and a subtherapeutic dose of BV (1 mg/kg, i.p.) with or without anti–PD-1 antibody (10 mg/kg, i.p. 2 and
7 days after PBMCs), showed enhanced immune-mediated tumor regression. C, Tumor volumes (AUC). Error bars indicate SEM.  P < 0.05;     P < 0.001
(one-way ANOVA followed by Dunnett’s multiple comparisons test). ANOVA, analysis of variance; AUC, area under the curve; BV, brentuximab vedotin; IgG,
immunoglobulin G; i.p., intraperitoneal injection; LCL, lymphoblastoid cell line; nivo, nivolumab; NT, not treated; PBMC, peripheral blood mononuclear cells;
PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; SEM, standard error of the mean; TIL, tumor-infiltrating lymphocyte.

direct tumor cell killing by brentuximab vedotin, priming and recruit- cells (44). MMAE is a synthetic derivative of the dolostatin 10 family
ment of T cells following ICD induction may help potentiate thera- of microtubule inhibitors with mechanistic similarities to vinca alkaloid
peutic blockade of PD-1/PD-L1 in CD30-expressing lymphomas. antineoplastic agents, vincristine and vinorelbine (45). Here, treatment
of CD30þ lymphoma cells with brentuximab vedotin or MMAE was
associated with induction of the three hallmarks of ICD: expression of
Discussion CRT on the cell surface, secretion of ATP, and extracellular release of
Failures in immunosurveillance can lead to cancer progression (1, 2). HMGB1 to a similar degree as the known ICD-inducing agent oxali-
Specifically, immunological ignorance of tumor-associated antigens, platin. Etoposide, a compound described not to induce ICD, was unable
immune exclusion, or active immunosuppression within the TME are to fulfill all three of these characteristics (a schematic of the proposed
key barriers to protective cancer immunity (1). Overcoming these mechanism is shown in Supplementary Fig. S6C). Unlike other known
barriers may be possible by eliciting tumor cell death and providing ICD-inducing agents that affect DNA replication/integrity (18, 12),
tumor-associated antigens in an inflammatory context (6). In contrast these results demonstrate that MMAE-mediated ICD is induced
with normal cellular apoptosis, ICD elicits the release of inflammatory through microtubule disruption and subsequent disorganization of the
DAMPs that promote APC activation and productive T-cell priming ER, leading to a potent ER stress response. Phagocytic uptake of
against tumor antigens (6). Therefore, induction of ICD by anticancer brentuximab vedotin–killed cells by APCs led to the production of
treatments is highly desirable to improve patient immune responses proinflammatory cytokines and efficiently primed the host immune
to cancer. system to mount a long-lived antitumor T-cell response. Overall, our
Brentuximab vedotin is an ADC that selectively delivers the highly results expand upon our understanding of brentuximab vedotin’s
potent microtubule inhibitor MMAE to CD30-expressing target multifaceted anticancer mechanism of action (Supplementary Fig. S6).

80 Mol Cancer Ther; 23(1) January 2024 MOLECULAR CANCER THERAPEUTICS


Brentuximab Vedotin and Immunogenic Cell Death

Brentuximab vedotin–induced innate cell activation was observed ICD hallmarks in ADC-treated cell lines (67, 68). Collectively, these
in an in vivo murine model by inducing high expression of hallmark findings support the hypothesis that induction of ICD is a vedotin
ICD cytokines. Blood biomarker analyses from a phase I/II study in ADC class effect and provide rationale for the clinical investigation of
patients with relapsed or refractory HL demonstrated a marked vedotin ADCs in combination with checkpoint inhibitors and other
increase in similar proinflammatory cytokines and chemokines fol- immunotherapies.
lowing treatment with brentuximab vedotin (33). These in-human
results validate the findings from the preclinical experiments inves- Authors’ Disclosures
tigating the potential for brentuximab vedotin to induce ICD. These studies were funded by Seagen Inc. R.A. Heiser, W. Zeng, M. Ulrich,
Antitumor immunity imparted by brentuximab vedotin–killed P. Younan, and R. Thurman are employees of Seagen and hold stocks and shares in
cells led to increased CD8þ T-cell infiltration and improved tumor Seagen. A.T. Cao, M.E. Anderson, M. Jonas, C.-L. Law, E.S. Trueblood, and S.J. Gardai
clearance in both murine and humanized tumor models. Given were employees of Seagen at the time of the studies. R.A. Heiser reports a patent
that clinical response rates to checkpoint inhibitors correlate with for US11299543B2 issued; as well as employment and being a stockholder of
the presence of CD8þ T cells in the TME (61), it was hypothesized Seagen Inc. A.T. Cao reports personal fees from Seagen during the conduct of the
study. P. Younan reports employment with Seagen. S.J. Gardai reports other support
that brentuximab vedotin–mediated ICD and the downstream from Seagen during the conduct of the study; as well as a patent for U.S. 201862668104
effects on T cells could be augmented by checkpoint inhibition. pending and U.S. 20200239585 pending. No other disclosures were reported by
Our models confirmed that brentuximab vedotin–mediated ICD the authors.
and resultant immune-mediated tumor rejection were enhanced by
PD-1 inhibition. Authors’ Contributions

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


These results provide a mechanistic rationale for investigating
R.A. Heiser: Conceptualization, investigation, writing–original draft, writing–
the antitumor response of brentuximab vedotin and checkpoint review and editing. A.T. Cao: Conceptualization, data curation, formal analysis,
inhibition as a novel regimen in CD30þ lymphomas. Previously investigation. W. Zeng: Investigation. M. Ulrich: Investigation. P. Younan: Inves-
published results of treatment with brentuximab vedotin with a tigation, methodology. M.E. Anderson: Investigation. E.S. Trueblood: Investigation.
PD-1 inhibitor (nivolumab) in patients have demonstrated prom- M. Jonas: Validation. R. Thurman: Writing–review and editing. C.-L. Law: Writing–
ising antitumor responses. In a phase I/II study in which 91 patients review and editing. S.J. Gardai: Conceptualization, investigation, writing–original
draft, writing–review and editing.
with relapsed or refractory HL were treated with brentuximab
vedotin plus nivolumab, the CR rate was 67% and the objective
response rate was 85%. Both the CR and objective response Acknowledgments
rates with brentuximab vedotin plus nivolumab were higher than Medical writing support was provided by Moamen Hammad, and editorial
previously observed with brentuximab vedotin or nivolumab support, including formatting, proofreading, and submission, was provided by
George Chappell and Travis Taylor, all of Scion, London, UK, supported by
alone (63). A similar study of brentuximab vedotin plus nivolumab Seagen according to Good Publication Practice guidelines (https://www.acpjour
in patients with relapsed or refractory non-Hodgkin lymphomas nals.org/doi/10.7326/M22-1460). Some of these results were presented at the
demonstrated an objective response rate of 70% and a 50% CR rate 2015, 2016, and 2017 American Association for Cancer Research (AACR) annual
in patients with mediastinal gray zone lymphoma (CHECKMATE meetings (49–51).
436; ref. 64). Ongoing studies are investigating the combination in
different populations; interim analyses of these studies have dem- The publication costs of this article were defrayed in part by the payment of
onstrated durable response rates in patients aged ≥60-years with publication fees. Therefore, and solely to indicate this fact, this article is hereby
marked “advertisement” in accordance with 18 USC section 1734.
classical HL (65) and high progression-free survival rates in patients
aged 9–30 years with relapsed or refractory classical HL (66).
The results of this study further demonstrate that the small- Note
Supplementary data for this article are available at Molecular Cancer Therapeutics
molecule MMAE itself is a potent driver of ICD irrespective of the
Online (http://mct.aacrjournals.org/).
anti-CD30 antibody backbone. Therefore, the ability to induce tar-
geted immune activation is likely intrinsic to other vedotin ADCs.
Indeed, preclinical studies with other vedotin ADCs (tisotumab Received March 3, 2023; revised August 7, 2023; accepted September 26, 2023;
vedotin and enfortumab vedotin) have demonstrated the presence of published first September 29, 2023.

References
1. Zhou J, Wang G, Chen Y, Wang H, Hua Y, Cai Z. Immunogenic cell death in 7. Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in
cancer therapy: present and emerging inducers. J Cell Mol Med 2019;23:4854–65. cancer and infectious disease. Nat Rev Immunol 2017;17:97–111.
2. de Souza AP, Bonorino C. Tumor immunosuppressive environment: effects on 8. Iyer SS, Pulskens WP, Sadler JJ, Butter LM, Teske GJ, Ulland TK, et al. Necrotic
tumor-specific and nontumor antigen immune responses. Expert Rev Antican- cells trigger a sterile inflammatory response through the Nlrp3 inflammasome.
cer Ther 2009;9:1317–32. Proc Natl Acad Sci USA 2009;106:20388–93.
3. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. 9. Gardai SJ, McPhillips KA, Frasch SC, Janssen WJ, Starefeldt A, Murphy-
Immunity 2013;39:1–10. Ullrich JE, et al. Cell-surface calreticulin initiates clearance of viable or
4. Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. apoptotic cells through trans-activation of LRP on the phagocyte. Cell 2005;
Induction of immunological tolerance by apoptotic cells requires caspase- 123:321–34.
dependent oxidation of high-mobility group box-1 protein. Immunity 2008; 10. Yu M, Wang H, Ding A, Golenbock DT, Latz E, Czura CJ, et al. HMGB1 signals
29:21–32. through toll-like receptor (TLR) 4 and TLR2. Shock 2006;26:174–9.
5. Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. 11. Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, et al.
Nat Immunol 2022;23:487–500. Nucleotides released by apoptotic cells act as a find-me signal to promote
6. Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, et al. phagocytic clearance. Nature 2009;461:282–6.
Consensus guidelines for the definition, detection and interpretation of immu- 12. Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer
nogenic cell death. J Immunother Cancer 2020;8:e000337. therapy. Annu Rev Immunol 2013;31:51–72.

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 81


Heiser et al.

13. Pitt JM, Kroemer G, Zitvogel L. Immunogenic and non-immunogenic cell 35. Hoimes CJ, Flaig TW, Milowsky MI, Friedlander TW, Bilen MA, Gupta S, et al.
death in the tumor microenvironment. Adv Exp Med Biol 2017;1036:65–79. Enfortumab vedotin plus pembrolizumab in previously untreated advanced
14. Tufi R, Panaretakis T, Bianchi K, Criollo A, Fazi B, Di Sano F, et al. Reduction of urothelial cancer. J Clin Oncol 2023;41:22–31.
endoplasmic reticulum Ca2þ levels favors plasma membrane surface exposure of 36. M€ uller P, Kreuzaler M, Khan T, Thommen DS, Martin K, Glatz K, et al.
calreticulin. Cell Death Differ 2008;15:274–82. Trastuzumab emtansine (T-DM1) renders HER2þ breast cancer highly suscep-
15. Panaretakis T, Kepp O, Brockmeier U, Tesniere A, Bjorklund AC, Chapman DC, tible to CTLA-4/PD-1 blockade. Sci Transl Med 2015;7:315ra188.
et al. Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell 37. M€ uller P, Martin K, Theurich S, Schreiner J, Savic S, Terszowski G, et al.
death. EMBO J 2009;28:578–90. Microtubule-depolymerizing agents used in antibody–drug conjugates induce
16. Kepp O, Menger L, Vacchelli E, Locher C, Adjemian S, Yamazaki T, et al. antitumor immunity by stimulation of dendritic cells. Cancer Immunol Res
Crosstalk between ER stress and immunogenic cell death. Cytokine Growth 2014;2:741–55.
Factor Rev 2013;24:311–8. 38. Martin K, M€ uller P, Schreiner J, Prince SS, Lardinois D, Heinzelmann-Schwarz
17. Rufo N, Yang Y, De Vleeschouwer S, Agostinis P. The “Yin and Yang” of VA, et al. The microtubule-depolymerizing agent ansamitocin P3 programs
unfolded protein response in cancer and immunogenic cell death. Cells 2022;11: dendritic cells toward enhanced antitumor immunity. Cancer Immunol Immun-
2899. other 2014;63:925–38.
18. Showalter A, Limaye A, Oyer JL, Igarashi R, Kittipatarin C, Copik AJ, et al. 39. Rios-Doria J, Harper J, Rothstein R, Wetzel L, Chesebrough J, Marrero A, et al.
Cytokines in immunogenic cell death: applications for cancer immunotherapy. Antibody–drug conjugates bearing pyrrolobenzodiazepine or tubulysin pay-
Cytokine 2017;97:123–32. loads are immunomodulatory and synergize with multiple immunotherapies.
19. Lebeau PF, Platko K, Byun JH, Austin RC. Calcium as a reliable marker for the Cancer Res 2017;77:2686–98.
quantitative assessment of endoplasmic reticulum stress in live cells. J Biol Chem 40. Montes de Oca R, Alavi AS, Vitali N, Bhattacharya S, Blackwell C, Patel K, et al.
2021;296:100779. Belantamab mafodotin (GSK2857916) drives immunogenic cell death and
20. Chaput N, De Botton S, Obeid M, Apetoh L, Ghiringhelli F, Panaretakis T, et al. immune-mediated antitumor responses in vivo. Mol Cancer Ther 2021;20:

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024


Molecular determinants of immunogenic cell death: surface exposure of calre- 1941–55.
ticulin makes the difference. J Mol Med 2007;85:1069–76. 41. Menger L, Vacchelli E, Adjemian S, Martins I, Ma Y, Shen S, et al. Cardiac
21. Obeid M, Panaretakis T, Joza N, Tufi R, Tesniere A, van Endert P, et al. glycosides exert anticancer effects by inducing immunogenic cell death.
Calreticulin exposure is required for the immunogenicity of gamma- Sci Transl Med 2012;4:143ra99.
irradiation and UVC light-induced apoptosis. Cell Death Differ 2007;14: 42. Sukkurwala AQ, Adjemian S, Senovilla L, Michaud M, Spaggiari S, Vacchelli E,
1848–50. et al. Screening of novel immunogenic cell death inducers within the NCI
22. Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, et al. Molecular mechanistic diversity set. Oncoimmunology 2014;3:e28473.
mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ 43. Seagen Inc. ADCETRIS US Full Prescribing Information. Updated November
2014;21:79–91. 2022. [Accessed October 9, 2023]. Available from: https://www.accessdata.fda.
23. Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, et al. Toll-like gov/drugsatfda_docs/label/2022/125388s106lbl.pdf.
receptor 4-dependent contribution of the immune system to anticancer che- 44. Yi JH, Kim SJ, Kim WS. Brentuximab vedotin: clinical updates and practical
motherapy and radiotherapy. Nat Med 2007;13:1050–9. guidance. Blood research 2017;52:243–53.
24. Tesniere A, Schlemmer F, Boige V, Kepp O, Martins I, Ghiringhelli F, et al. 45. Gauzy-Lazo L, Sassoon I, Brun MP. Advances in antibody–drug conjugate
Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene design: current clinical landscape and future innovations. SLAS Discov 2020;
2010;29:482–91. 25:843–68.
25. Garg AD, Krysko DV, Verfaillie T, Kaczmarek A, Ferreira GB, Marysael T, et al. 46. Jain N, Smith SW, Ghone S, Tomczuk B. Current ADC linker chemistry.
A novel pathway combining calreticulin exposure and ATP secretion in immu- Pharm Res 2015;32:3526–40.
nogenic cancer cell death. Embo j 2012;31:1062–79. 47. Sutherland MS, Sanderson RJ, Gordon KA, Andreyka J, Cerveny CG, Yu C, et al.
26. Michaud M, Sukkurwala AQ, Di Sano F, Zitvogel L, Kepp O, Kroemer G. Lysosomal trafficking and cysteine protease metabolism confer target-specific
Synthetic induction of immunogenic cell death by genetic stimulation of cytotoxicity by peptide-linked anti-CD30–Auristatin conjugates. J Biol Chem
endoplasmic reticulum stress. Oncoimmunology 2014;3:e28276. 2006;281:10540–7.
27. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, et al. 48. Barreca M, Stathis A, Barraja P, Bertoni F. An overview on anti-tubulin
Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med agents for the treatment of lymphoma patients. Pharmacol Ther 2020;211:
2007;13:54–61. 107552.
28. Martins I, Kepp O, Schlemmer F, Adjemian S, Tailler M, Shen S, et al. Restoration 49. Gardai SJ, Epp A, Law C-L. Abstract 2469: Brentuximab vedotin-mediated
of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic immunogenic cell death. Cancer Res 2015;75:2469.
reticulum stress. Oncogene 2011;30:1147–58. 50. Cao A, Heiser R, Law C-L, Gardai SJ. Abstract 4914: Auristatin-based antibody–
29. Verma R, Foster RE, Horgan K, Mounsey K, Nixon H, Smalle N, et al. drug conjugates activate multiple ER stress response pathways resulting in
Lymphocyte depletion and repopulation after chemotherapy for primary breast immunogenic cell death and amplified T-cell responses. Cancer Res 2016;76:
cancer. Breast Cancer Res 2016;18:10. 4914.
30. Mackall CL, Fleisher TA, Brown MR, Andrich MP, Chen CC, Feuerstein IM, et al. 51. Cao AT, Law C-L, Gardai SJ, Heiser RA. Abstract 5588: Brentuximabvedotin-
Distinctions between CD8þ and CD4þ T-cell regenerative pathways result in driven immunogenic cell death enhances antitumor immune responses, and is
prolonged T-cell subset imbalance after intensive chemotherapy. Blood 1997;89: potentiated by PD-1 inhibition in vivo. Cancer Res 2017;77:5588.
3700–7. 52. Ma Y, Adjemian S, Yang H, Catani JP, Hannani D, Martins I, et al. ATP-
31. Kotsakis A, Sarra E, Peraki M, Koukourakis M, Apostolaki S, Souglakos J, et al. dependent recruitment, survival and differentiation of dendritic cell precursors
Docetaxel-induced lymphopenia in patients with solid tumors: a prospective in the tumor bed after anticancer chemotherapy. Oncoimmunology 2013;2:
phenotypic analysis. Cancer 2000;89:1380–6. e24568.
32. Fagnoni FF, Lozza L, Zibera C, Zambelli A, Ponchio L, Gibelli N, et al. T- 53. Terasaki M, Chen LB, Fujiwara K. Microtubules and the endoplasmic reticulum
cell dynamics after high-dose chemotherapy in adults: elucidation of the are highly interdependent structures. J Cell Biol 1986;103:1557–68.
elusive CD8þ subset reveals multiple homeostatic T-cell compartments 54. Schwarz DS, Blower MD. The endoplasmic reticulum: structure, function, and
with distinct implications for immune competence. Immunology 2002;106: response to cellular signaling. Cell Mol Life Sci 2016;73:79–94.
27–37. 55. Klopfenstein DR, Kappeler F, Hauri HP. A novel direct interaction of endo-
33. Herrera AF, Moskowitz AJ, Bartlett NL, Vose JM, Ramchandren R, Feldman TA, plasmic reticulum with microtubules. Embo j 1998;17:6168–77.
et al. Interim results of brentuximab vedotin in combination with nivolumab 56. Ko P, Choi JH, Song S, Keum S, Jeong J, Hwang YE, et al. Microtubule acetylation
in patients with relapsed or refractory Hodgkin lymphoma. Blood 2018;131: controls MDA-MB-231 breast cancer cell invasion through the modulation of
1183–94. endoplasmic reticulum stress. Int J Mol Sci 2021;22:6018.
34. Nicol o E, Giugliano F, Ascione L, Tarantino P, Corti C, Tolaney SM, et al. 57. Dominguez-Bautista JA, Klinkenberg M, Brehm N, Subramaniam M, Kern B,
Combining antibody–drug conjugates with immunotherapy in solid tumors: Roeper J, et al. Loss of lysosome-associated membrane protein 3 (LAMP3)
current landscape and future perspectives. Cancer Treat Rev 2022;106: enhances cellular vulnerability against proteasomal inhibition. Eur J Cell Biol
102395. 2015;94:148–61.

82 Mol Cancer Ther; 23(1) January 2024 MOLECULAR CANCER THERAPEUTICS


Brentuximab Vedotin and Immunogenic Cell Death

58. Gombault A, Baron L, Couillin I. ATP release and purinergic signaling in NLRP3 mediastinal gray zone lymphoma: primary efficacy and safety analysis of the
inflammasome activation. Front Immunol 2012;3:414. phase 2 CheckMate 436 study. Blood 2020;136:44–5.
59. Wang Y, Martins I, Ma Y, Kepp O, Galluzzi L, Kroemer G. Autophagy- 65. Yasenchak CA, Bordoni R, Patel-Donnelly D, Larson T, Goldschmidt J,
dependent ATP release from dying cells via lysosomal exocytosis. Autophagy Boccia RV, et al. Frontline brentuximab vedotin as monotherapy or
2013;9:1624–5. in combination for older Hodgkin lymphoma patients. Blood 2020;136:
60. Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, et al. 18–9.
Consensus guidelines for the detection of immunogenic cell death. Oncoimmu- 66. Cole P, Mauz-K€orholz C, Mascarin M, Michel G, Cooper S, Beishuizen A, et al.
nology 2014;3:e955691. Nivolumab and brentuximab vedotin (BV)-based, response-adapted treatment
61. Khagi Y, Kurzrock R, Patel SP. Next-generation predictive biomarkers for in children, adolescents, and young adults (CAYA) with standard-risk relapsed/
immune checkpoint inhibition. Cancer Metastasis Rev 2017;36:179–90. refractory classical Hodgkin lymphoma (R/R cHL): primary analysis. J Clin
62. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, Oncol 2020;38:8013.
et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 67. Gray E, Hensley K, Allred S, Trueblood E, Gosink J, Thurman R, et al. 617
expression. Cell Rep 2017;19:1189–201. Tisotumab vedotin shows immunomodulatory activity through induction of
63. Advani R, Moskowitz AJ, Bartlett NL, Vose J, Ramchandren R, Feldman T, et al. immunogenic cell death. J Immunother Cancer 2020;8:A653.
Brentuximab vedotin in combination with nivolumab in relapsed or refractory 68. Liu BA, Olson D, Snead K, Gosink J, Tenn E-M, Zaval M, et al. Abstract 5581:
Hodgkin lymphoma: 3-year study results. Blood 2021;138:427–38. enfortumab vedotin, an anti–Nectin-4 ADC demonstrates bystander cell killing
64. Santoro A, Moskowitz AJ, Ferrari S, Carlo-Stella C, Fanale MA, Francis S, and immunogenic cell death antitumor activity mechanisms of action in
et al. Nivolumab combined with brentuximab vedotin for relapsed/refractory urothelial cancers. Cancer Res 2020;80:5581.

Downloaded from http://aacrjournals.org/mct/article-pdf/23/1/68/3397457/68.pdf by guest on 13 February 2024

AACRJournals.org Mol Cancer Ther; 23(1) January 2024 83

You might also like