Play all audios:
ABSTRACT Tumor-associated neutrophils (TANs) actively interact with antibody-drug conjugates (ADCs) within the tumor microenvironment (TME), though the detailed mechanisms governing their
response to ADC treatment remain to be fully elucidated. Herein, we explored how ICAM1-targeted ADCs affect TAN dynamics in preclinical models of cervical cancer. We discovered that I-DXd,
our in-house ADC targeting cervical cancer, effectively eliminates tumor cells through specific antigen recognition while concurrently depleting pro-tumor VEGFA + TANs via Fcγ
receptor-mediated phagocytosis. This dual action promotes an immunologically favorable TME. Through comprehensive preclinical studies, we established a foundational understanding of the
synergistic benefits of combining I-DXd treatment with PD-1 immune checkpoint inhibition, thereby opening new avenues for therapeutic intervention in advanced cervical cancer. SIMILAR
CONTENT BEING VIEWED BY OTHERS EFFECTIVE EXTRACELLULAR PAYLOAD RELEASE AND IMMUNOMODULATORY INTERACTIONS GOVERN THE THERAPEUTIC EFFECT OF TRASTUZUMAB DERUXTECAN (T-DXD) Article Open access
02 April 2025 COPPER CHELATION REDIRECTS NEUTROPHIL FUNCTION TO ENHANCE ANTI-GD2 ANTIBODY THERAPY IN NEUROBLASTOMA Article Open access 12 December 2024 POTENTIATION OF IMMUNE CHECKPOINT
BLOCKADE WITH A PH-SENSITIZER AS MONITORED IN TWO PRE-CLINICAL TUMOR MODELS WITH ACIDOCEST MRI Article 24 February 2025 INTRODUCTION Cervical cancer remains a lethal female cancer accounting
for over 0.34 million annual mortalities globally [1, 2]. The clinical outcomes for patients with gynecologic malignancies have not been improved over the past four decades, largely due to
the lack of effective treatment modalities [2,3,4,5]. Given the lack of second-line therapeutic agents for advanced and metastatic cervical cancer, the 5-year survival rate has plummeted
from 92% to 15% and 20% for patients with late-stage cervical cancer [6,7,8]. Antibody-drug conjugates (ADCs) have emerged as revolutionary targeted therapeutics for various refractory and
late-stage solid tumors including cervical and ovarian cancers [9,10,11]. Tisotumab Vedotin, a tissue-factor-targeted ADC, is the first clinically-approved ADC for advanced cervical cancers.
However, in two recent large cohorts of Phase II clinical trials (innovaTV204 and innovaTV207), Tisotumab Vedotin yielded modest therapeutic benefits of overall response rates (ORR) of 24%
in patients with advanced cervical cancer and 16% of other solid tumors [12], which is much less effective than Trastuzumab Deruxtecan (ORR: 50%), another HER2-targeted ADC with different
linker and payload design, in the DESTINY-PanTumor02 Phase II trial [13]. Therefore, a deeper understanding of the drug mechanism(s) of ADCs is urgently needed to guide the development of
novel ADC agents. Our previous drug mechanism study of ADC chemical structures revealed that the linker and payload combination of Tisotumab Vedotin (MC-VC-PAB-MMAE) conferred significant
differences in tumor immune microenvironment in comparison with that of Trastuzumab Deruxtecan (MC-GGFG-DXd), resulting in suboptimal bystander killing effects and therapeutic efficacies in
mice [14, 15]. In particular, we found that tumor-associated neutrophils (TANs) were identified as the most affected immune cells by ADCs among innate and adaptive immune cells within the
TME. Neutrophils are the most abundant innate immune cells in the human immune system and TANs exhibit functional heterogeneity and plasticity, including pro-tumor and anti-tumor phenotypes
in TME [16, 17]. In advanced tumors, TANs commonly are polarized into immunosuppressive phenotypes due to stimuli by tumor cells and TME components [18, 19]. However, the underlying
biomechanism(s) of how ADCs affect TANs and subsequent ignited immune responses in TME have yet to be elucidated. Herein, we identified ICAM1 as an optimal ADC target for cervical cancer and
used it to construct two ADCs using the same linker and payload combinations of Tisotumab Vedotin and Trastuzumab Deruxtecan, and comprehensively explored their impacts on TAN elimination
and polarization during ADC treatment. These analyses revealed that TANs effectively phagocytose intact ADC macromolecules and facilitated Cathepsin B-mediated linker cleavage, exerting
potent bystander killing activity in eliminating VEGFA+ TANs, which in turn, ignites type I interferon (IFN) signaling cascade and increases CD8 + T cell recruitment. These findings suggest
that the immunoregulatory functions of ADCs could offer an opportunity to potentiate PD-1 immunotherapy in cervical cancer and other solid tumors. RESULTS IDENTIFICATION OF ICAM1 AS A NOVEL
ADC TARGET FOR CERVICAL CANCER We first performed an unbiased and quantitative screening of a library of 67 cell membrane proteins on a panel of three cervical cancer cell lines (SiHa,
CaSki, ME180) and normal human cervical epithelial HcerEpic cells. 4 of 67 proteins (ICAM1, CD49b, CD73, and EGFR) were identified as consistently upregulated in all cervical cancer cell
lines (Fig. 1a). Notably, ICAM1, an inflammation-related cell membrane protein, showed potential as a specific molecular target candidate for cervical cancer. By querying the Human Protein
Atlas database (https://www.proteinatlas.org), CD49b, CD73, and EGFR are highly expressed in normal tissues due to their poor specificity. CD49b was expressed in 71.11% (32/45) of normal
tissues, CD73 in 86.05% (37/43), and EGFR in 46.67% (21/45), which may cause more on-target-off-tumor toxicities in contrast to ICAM1 in 19.15% (9/47). Therefore, ICAM1 was selected as the
target candidate for further investigation. To validate its clinical relativity, we queried the GEPIA database for ICAM1 expression in a total of 306 patients with cervical cancer and found
that the level of ICAM1 mRNA was significantly upregulated in patients with cervical cancer (Fig. 1b). To validate this finding at the protein level, we performed immunohistochemical (IHC)
staining of ICAM1 on a microarray of 111 human cervical tumor tissues including 107 squamous cell carcinoma and 4 adenocarcinoma (Fig. 1c). The IHC staining of ICAM1 expression shows
heterogeneous patterns in cervical tumor tissues. Regions with low or absent ICAM1 expression primarily consist of regions of necrosis and non-malignant stromal cells, including fibroblasts,
and infiltrating immune cells such as lymphocytes and macrophages, which typically do not express high levels of ICAM1. This contrasts with the areas where tumor cells with high ICAM1
expression are clustered, exhibiting stronger staining. Additionally, the whole tissue section staining (Supplementary Fig. 1) confirms this pattern, showing ICAM1 is predominantly expressed
in tumor cell regions while being low or absent in surrounding stromal and non-cancerous areas. Importantly, the ICAM1 positive rate in cervical tumor tissues was determined as 24.32%
(27/111). Among them, 26.87% (18/67) were determined as ICAM1-high expression and 21.05% (8/38) as ICAM1-low expression. In comparison, ICAM1 expression is completely absent in 25
paracancerous normal human tissues (Fig. 1d). The IHC staining scoring criteria of ICAM1 are defined as shown in the supplementary information (Supplementary Fig. 2) and the IHC method
section. Moreover, high ICAM1 expression was associated with a worse prognosis in patients with cervical cancer (Fig. 1e). We have previously assessed the ICAM1 expression in a cohort of 144
normal human tissues of 20 different organs by IHC showing low or undetectable ICAM1 expression in most organs [20]. We next quantified ICAM1 overexpression in human cervical cancer cells
(SiHa and CaSki) by flow cytometry and immunofluorescence staining, which are 80- and 106-fold higher than those of non-neoplastic HcerEpic and 293T cells, respectively (Fig. 1f, g).
Moreover, immunofluorescence staining shows that ICAM1 was localized on the plasma membrane of SiHa and CaSki cells (Fig. 1h), which is readily accessible for antibody-based drugs. Given
that cell entry is a critical factor affecting the therapeutic efficacy of ADC [21, 22], we evaluated the internalization rate of ICAM1 antibodies in human cervical cancer cells. As shown in
Fig. 1i, j, fluorophore phycoerythrin (PE) -conjugated ICAM1 antibodies rapidly entered SiHa and CaSki cells, reaching a plateaued internalization rate of approximately 30% within 120 min.
In addition, the cell viability test shows that the ICAM1 antibody did not affect the cell viability during this internalization process (Supplenmentary Fig. 3). Furthermore, the
internalized ICAM1 antibodies were observed to colocalize with the lysosomes (LAMP1+, Supplenmentary Fig. 4) of cervical cancer cells, confirming that the internalized antibody traffics to
intracellular compartments, validating the internalization process of ICAM1 in these cells. Overall, the above findings strongly support ICAM1 as a promising ADC target for cervical cancer
therapy. GENERATION AND CHARACTERIZATION OF ICAM1-ADCS To find the optimal ADC construct for cervical cancer therapy, we compared the antitumor efficacy of two ICAM1-ADCs of different
mechanisms of action (MoAs) previously developed by us. The two ICAM1-ADCs were constructed by conjugating ICAM1 monoclonal antibodies with two clinically approved linker and payload
combinations from Tisotumab Vedotin and Trastuzumab Deruxtecan, MC-VC-MMAE and MC-GGFG-DXd. The obtained ADCs were termed ICAM1-MMAE (I-MMAE) and ICAM1-DXd (I-DXd) based on their cytotoxic
payloads. I-MMAE features a microtubule inhibitor MMAE payload whereas I-DXd uses DNA topoisomerase inhibitor DXd as payload (Fig. 2a, b), which could ablate cervical cancer cells via
different MoAs. Both I-MMAE and I-DXd utilized cathepsin B enzyme-cleavable peptide linkers, providing a bystander-killing effect within the TME. The drug-to-antibody ratios (DARs) of I-MMAE
and I-DXd were fine-tuned to 4 and 8 (Fig. 2a, b), respectively, in consistency with Tisotumab Vedotin and Trastuzumab Deruxtecan of the same linker and payload combinations [14, 23]. We
next determined the in vitro cytotoxicity of two ICAM1-ADC constructs in a panel of cervical cancer (SiHa and CaSki) and non-neoplastic (HcerEpic and 293T) cell lines (Supplementary Fig. 5).
The half maximum inhibitory concentration (IC50) of I-MMAE was determined as 0.1198 µg/ml (SiHa) and 1.149 µg/ml (CaSki), respectively. For I-DXd with a less cytotoxic payload DXd, its IC50
was approximately 10 µg/ml for SiHa and CaSki cells. Notably, both ICAM1-ADC constructs did not induce any cytotoxicity on normal human cervical cells (HcerEpic). Cisplatin (Cis) and
Paclitaxel (Pac) are two of the conventional first-line chemo drugs for cervical cancer, which were used as positive controls. As shown in Fig. S5, Cis and Pac ubiquitously ablate both
cervical cancer and non-neoplastic cells without any tumor-selectivity. These in vitro efficacy results support that two ICAM1-ADCs are potent in cervical cancer cells with high tumor
selectivity, warranting further investigation in the in vivo cervical tumor models. ICAM1-ADCS POTENTLY SUPPRESS CERVICAL TUMORS IN VIVO Given the lack of second-line therapeutic agents for
advanced and metastatic cervical cancer, the five-year survival rate has plummeted from 92% to 15% and 20% for patients with late-stage cervical cancer [6,7,8]. Thus, this study evaluated
the therapeutic potential of ICAM1-ADCs in both standard and late-stage cervical tumor models. ICAM1-positive SiHa cells were utilized to establish the standard and late-stage models of
human cervical tumors in nude mice (Fig. 2c). In the standard setting, the established protocol entailed administering I-MMAE and I-DXd through the caudal vein once tumor volume reached
50–80 mm3. The first-line chemotherapy drug Cis was utilized as a positive control, while ICAM1 antibody (I Ab) and PBS served as the negative controls. The recommended dosage [15, 24] for
I-Dxd, I-MMAE, and Cis is 5 mg/kg, administered twice, on day 0 and day 7, respectively (Fig. 2c). After treatment, the tumor in the control group treated with PBS increased linearly while
both I-MMAE and I-DXd groups showed a significant reduction in cervical tumor growth (Fig. 2d, c). Notably, there was no significant difference between the efficacies of the two ADCs in the
standard setting (Fig. 2c). In comparison, Cis treatment only showed a moderate inhibitory effect on cervical tumors. Quantified terminal tumor weights confirmed that the antitumor effects
of two ICAM1 ADCs are significantly more potent than Cis (44.32%) at the same dose with their tumor inhibition rates determined as 85.80% (I-MMAE) and 91.38% (I-DXd), noting that ICAM1
antibody alone had no obvious inhibitory effect on the cervical tumor (Fig. 2f). In the late-stage setting [25], ICAM1 ADC treatment was initiated when the tumor volume reached approximately
500 mm3. Consistently, both I-MMAE and I-DXd showed potent tumor inhibitory activity in comparison with other control groups, with I-DXd exhibiting the strongest antitumor effect (Fig. 2).
Their tumor inhibition rates in the late-stage setting were determined as 50.10% (I-MMAE) and 68.64% (I-DXd), substantially more potent than 41.49% of the Cis group (Fig. 2j). We further
evaluated animal survival by Kaplan-Mier analysis, and in comparison with the PBS group, two ICAM1-ADC groups significantly improved their prognosis, which was consistent with their
antitumor effects (Fig. 2k). While we recognize that this categorization of standard and late-stage cervical tumor models does not capture inherent molecular differences in cervical cancer
subtypes, it allows us to assess the therapeutic efficacy of ADCs at varying tumor burdens, which is relevant for understanding treatment response in clinical settings with different tumor
sizes. Collectively, the I-DXd construct exhibits consistently superior antitumor activity with improved prognosis in both standard and late-stage models, which is recommended for further
cervical cancer therapy studies. ICAM1 ADCS ARE SAFE AND WELL-TOLERATED IN MICE We assessed the toxicity of I-MMAE and I-DXd in multiple human cervical cell line-derived xenograft (CDX)
models via blood biochemistry. First, we observed no significant bodyweight changes upon ICAM1 ADC treatment in both standard and late-stage settings of SiHa tumors (Fig. 2g, i). We further
analyzed a panel of serum biomarkers related to liver and renal functions and immunogenicity [26, 27]. As shown in Fig. 2n, I-MMAE and I-DXd treatments did not alter the levels of ALT
(glutamic pyruvic transaminase), AST (glutamic oxaloacetic transaminase), ALP (alkaline phosphatase), CHE (cholinesterase), and TBIL (total bilirubin) related to liver functions. Similar
trends were also observed in renal function and immunogenicity biomarkers including CREA (creatinine), UREA (urine urea nitrogen), UA (uric acid), and HCP (hypersensitive C-reactive protein)
(Fig. 2m). Collectively, the above findings support that the I-MMAE and I-DXd single agents are safe and well tolerated in both standard and late-stage mice settings. I-DXD DOWNREGULATES
NEUTROPHIL ACTIVATION IN CERVICAL TME AND ABLATES PRO-TUMOR TANS We previously reported that TANs are the most affected immune cells in TME by Trastuzumab Deruxtecan, a clinical
HER2-targeted ADC with the same linker and payload combination with I-DXd [15]. In this study, we specifically investigated the detailed immunoregulatory functions of I-DXd on TANs in
cervical TME. Neutrophils are the most numerous innate immune cells in TME and have the dual nature of pro-tumor and anti-tumor during tumor progression [16, 17]. We first performed the
whole transcriptomic analysis of ICAM1-ADC treated cervical tumors from the late-stage tumor models via RNA-sequencing (RNA-seq). Figure 3a shows the top 1000 differentially expressed genes
(DEGs) of PBS, ICAM1 antibody, I-MMAE, and I-DXd groups, and the I-DXd group exhibited a significantly changed phenotype in comparison with other groups. Gene Ontology (GO) enrichment
analysis revealed that I-DXd significantly downregulated four pathways associated with neutrophil activation different from I-MMAE (Fig. 3b), indicating that the neutrophil activation
process was suppressed in cervical TME after I-DXd treatment. Next, we performed an IHC staining on the ICAM1 ADC-treated cervical tumors harvested from the late-stage model. TANs were
labeled by Ly6G and pro-tumor subsets of TANs were identified by MPO, an established biomarker for tumor-promoting neutrophils [28, 29]. IHC staining results (Fig. 3c) showed that I-MMAE and
I-DXd potently eliminated tumor-infiltrating TANs in TME at both total TANs (Ly6G+) and pro-tumor TANs (MPO+) levels, and I-DXd reached a superior TAN elimination rate than I-MMAE, as high
as 75% for total TANs and 85% for pro-tumor TANs, respectively (Fig. 3d). We validated this critical finding in an independent flow cytometry assay (Fig. 3e) and found that CD45 + CD11b +
Ly6G + TANs accounted for 92.92% of the CD11b+ myeloid cells in the PBS group, while it was significantly reduced to 22.68% and 12.09% of the myeloid cells in the I-MMAE and I-DXd groups,
respectively (Fig. 3f). These findings strongly support that I-MMAE and I-DXd effectively eliminate pro-tumor TANs (Ly6G + MPO + TANs) in cervical TME. In addition, the heterogeneous
staining signals of Ly6G and MPO in the PBS-treated group reflect the variable distribution of neutrophils within the tumor microenvironment (Fig. 3c and Supplementary Fig. 6). This pattern
indicates that neutrophils are not uniformly distributed across the tumor tissue, with certain regions showing a higher density of Ly6G+ and MPO+ cells. This variability is commonly observed
in tumors, where factors contributing to this distribution pattern include tumor architecture, hypoxia, and intra-tumoral chemokine gradients within the tumor microenvironment [16, 30]. In
areas with higher neutrophil infiltration, Ly6G, and MPO staining intensities are stronger, likely due to the recruitment of neutrophils in response to local tumor cues. Conversely, regions
with lower staining indicate areas where neutrophil infiltration is minimal or absent. Such heterogeneous distribution aligns with the dynamic and complex nature of the tumor
microenvironment, where immune cell populations often exhibit spatial variability. I-DXD EFFECTIVELY ABLATES VEGFA + TANS AND BOOSTS ANTITUMOR IMMUNITY Previous studies have identified 20
cytokine biomarkers associated with TAN functionality in the TME [30]. By comparing the gene scores of differentially expressed genes between I-DXd and PBS control groups in TAN-related gene
sets, we identified 12 significant target genes, among which 8 genes were significantly downregulated and 4 genes upregulated (Fig. 4a). Some studies defined VEGFA, CCL2, CCL28, and CSF3,
which are related to TAN, as protumor genes in TME, whereas ARG1, MMP9, and CXCL2 were defined as antitumor genes [31, 32]. We further performed a more detailed TAN-associated gene analysis
and found that I-DXd significantly downregulates protumor TAN genes (e.g., VEGFA, CCL2, CCL28, and CSF3) (Fig. 4b) and upregulated antitumor TAN genes (e.g., ARG1, MMP9, and CXCL2) (Fig.
4c). Thus, we can reasonably hypothesize that TANs with these 12 significant TAN-related target genes are defined as VEGFA + TANs, which appear to be an immunosuppressive population in the
advanced cervical cancer TME. To assess the clinical relevance of VEGFA+ TANs, gene set variation analysis (GSVA) was employed, demonstrating significant enrichment of these TANs in tumor
samples over normal tissues within the GSE63514 database (Fig. 4d, e). Furthermore, a high GSVA score for VEGFA+ TANs correlated with a poorer clinical prognosis in cervical cancer patients
according to the TCGA database (Fig. 4f). In correlation, a recent TAN profiling study also reported that the VEGFA + SPP1+ subset was associated with worse clinical outcomes in 225 samples
from 145 patients of 17 solid tumors including cervical cancer [33, 34]. We next used EPIC (https://gfellerlab.shinyapps.io/EPIC_1-1), a tool for predicting TME cell composition, to
investigate changes in the proportion of immune cells in the TME of cancer patients between high and low TAN-related GSVA score groups in the TCGA database. Differences in immune cell
proportions were observed between patients with high and low TAN-related GSVA scores, suggesting a link between lower scores and improved clinical outcomes, possibly due to enhanced CD8 + T
cell activation (Fig. 4g). These findings further support the concept that VEGFA+TANs, as a whole, exhibit a tumor-promoting function of immunosuppression in the cervical cancer clinical
database, which is reversed or eliminated by I-DXd treatment. We next investigated the interaction between I-DXd and TANs. Given TANs are innate immune cells with phagocytic functions of
foreign microparticles including bacteria and viruses [17], we isolated mouse TANs from human cervical tumors (SiHa) and co-incubated them with fluorophore Cy5.5-labeled I-DXd and I-MMAE in
comparison with circulating neutrophils (quiescent) derived from mouse peripheral blood. As a result, mouse TANs showed effective phagocytosis of both ICAM1-ADC constructs, whereas quiescent
neutrophils did not show obvious phagocytosis of these ADCs (Fig. 4h). Because the ICAM1 antibody of these ADC constructs only binds human antigens without mouse reactivity, the TAN
phagocytosis of ICAM1-ADCs is independent of ICAM1 antigen but dependent on the FcγR engagement [35,36,37]. Strikingly, our immunoblot results (Fig. 4i and Supplementary Fig. 7) revealed
that mouse TANs (T1 and T2) also express high levels of Cathepsin B, the major protease cleaving peptide linkers of ICAM1-ADCs, in comparison with peripheral neutrophils (P1 and P2),
indicating that TANs could cleave intact ICAM1-ADCs into small molecular payloads and subsequently exert the bystander killing effect, as well as antigen-positive tumor cells in TME, leading
to enhanced an amplified antitumor activity of ICAM1-ADCs. Collectively, these findings elicit that ICAM1-ADCs effectively ablate protumor VEGFA+ TANs in TME through antigen-independent
phagocytosis and TAN-mediated bystander-killing effects. I-DXD POTENTIATES PD-1 IMMUNOTHERAPY VIA TYPE I IFN RESPONSES We investigated the immunoregulatory functions of ADC-mediated TAN
elimination on tumor immune microenvironment. Based on our transcriptome analysis on I-DXd-treated tumors, the GSEA map shows an enriched interferon (IFN) alpha-related pathway, an
established type I IFN immune response (Fig. 5a). Upregulation of IFN signaling in the TME is closely associated with activation of adaptive immunity and antitumor immune stimulation
[38,39,40]. In Fig. 5b, our DEG analysis of IFN signal pathway-related genes showed significant upregulation of a panel of immunoreactive genes including OASL, GBP4, SAMD9L, IFIT3, RSAD2,
IFI44, STAT2, MX1, and IFIF27 upon I-DXd treatment (Fig. 5c, d), indicating that I-DXd effectively ameliorates tumor immune microenvironment by activating type I IFN responses. Since type I
IFN signaling has an essential role in the full-blown efficacy of many immunotherapy agents, especially PD1 immune checkpoint blockade therapy [41], we postulated that I-DXd may potentiate
PD1 immunotherapy by boosting antitumor immunity through TAN elimination. Additionally, to confirm that immune activation (such as the IFN signaling pathway) is indeed induced by I-DXd
treatment rather than ICAM1 antibody, we analyzed the potential signaling transductions in human cervical tumors by ICAM1 neutralizing antibody using RNA transcriptomic analyses. GO analysis
(Supplementary Fig. 8) demonstrates the top 20 up- and down-regulated signaling pathways initiated by ICAM1 neutralizing antibody treatment. The regulation of cellular response to the
hypoxia pathway was found as the most significantly downregulated signaling pathway upon ICAM1 neutralizing antibody treatment, but further studies are needed to characterize the specific
pathways activated in cervical cancer cells. Intriguingly, others have recently reported that ICAM1 neutralizing antibodies effectively inhibit non-small cell lung tumor growth by activating
Caspases 3 and 9 and inhibiting AKT and ERK1/2 pathways [42]. In addition, ICAM1 neutralizing antibodies have also been found to inhibit SRC and STAT3 pathways and inhibit colon tumor
metastasis and angiogenesis [43]. More importantly, we also examined the I-MMAE on the phenotypes of neutrophils in the same TAN- and IFN-related gene sets (Supplementary Fig. 9). Compared
to the I-DXd treatment, the protumoral genes related to TAN are slightly downregulated in the I-MMAE treatment, especially VEGFA, CSF3, CCL28, and CCL2 (Supplementary Fig. 9a, b). Although
the panel of immunoreactive genes (IFN-related genes) is slightly upregulated by I-MMAE treatment (Supplementary Fig. 9c, d), I-DXd is more effective in activating neutrophil-related immune
responses than I-MMAE as observed in Fig. 3b. Subsequently, we focused on the effects of I-DXd and its combo therapy on immune activation due to its unique mechanism and promising
preclinical results. I-DXD COMBINED WITH PD1 ANTIBODY INDUCES CD8 + T CELL RECRUITMENT AND ENHANCES THE ANTI-TUMOR EFFECT We next evaluated the synergistic effects of I-DXd in combination
with PD1 immunotherapy in an immunocompetent murine tumor model. An immunocompetent Balb/c mouse model was established by using murine colorectal carcinoma CT26 cells stably expressing human
ICAM1 (CT26-hICAM1) due to low tumor formation rates of mouse cervical cancer cell lines. I-DXd, anti-mouse PD-1 monoclonal antibody, or their combination were intravenously administered in
comparison with the PBS group (Fig. 6a). Our results show that I-DXd or PD-1 monotherapy yielded moderate tumor inhibitory activities whereas the combo group exhibited the highest antitumor
effect, significantly better than I-DXd or PD-1 monotherapy (Fig. 6b). We further evaluated the prognosis of I-DXd and PD-1 combination therapy via Kaplan-Mier analysis. Consistently, I-DXd
and Combo groups demonstrated the most favorable prognosis with 83.3% (5/6) mice surviving. In comparison, PBS and PD-1 monotherapy groups had no survivors at the end of the experiment
(Fig. 6c). Considering I-DXd and PD-1 combination therapy may induce additive toxicities of each agent, in addition to bodyweight measurement (Fig. 6d), we also performed biochemistry
analyses on mouse blood samples collected from immunocompetent Balb/C mice receiving I-DXd/PD-1 combination therapy. As shown in Fig. 6e, f, no obvious changes were observed in either
I-DXd/PD-1 monotherapy or the Combo group. Given adaptive immunity is pivotal in PD-1 immunotherapy, we investigated the impact of I-DXd and PD-1 combination therapy on CD4/CD8 + T cell
recruitment by IHC staining and flow cytometry (Fig. 6g, h). We observed significant increases in CD8 + T cells and decreases in CD4 + T cells in I-DXd, PD-1, and the Combo group in
comparison with the PBS group (Fig. 6i–k). Collectively, our findings support that I-DXd works in synergy with PD-1 immunotherapy via type I IFN responses and subsequently improved CD4/CD8 T
cell ratios in TME. NON-INVASIVE IMAGING OF ICAM1+ CERVICAL TUMOR BY MRI Given ADC efficacy directly relies on the levels of tumoral antigen expression, tissue biopsy via vaginal endoscopy
is frequently required before ADC treatment to assess target adequacy in clinical practice. However, this approach is invasive and potentially causes bleeding and increases the risk of
metastasis [44,45,46]. We previously reported an MRI-based molecular probe by conjugating ICAM1 antibody conjugated with a magnetic resonance T1 contrast agent Gadolinium (I-Gd) [47, 48].
Here we utilized I-Gd to non-invasively quantify tumoral antigen expression of ICAM1 in the CDX model of human cervical tumor (SiHa) via MRI. At pre- and post-I-Gd injections, we performed
T1-weighted MR imaging to quantify MRI signal changes at tumor regions as a function of Gd accumulation from administered probes (Fig. 7a, b). In comparison with the non-targeting IgG-Gd
group, I-Gd yielded a 138% significantly greater increase in MRI signal in comparison with its background, while IgG-Gd only resulted in a 38% increase (Fig. 7c). The results indicate that
the I-Gd MRI probe can accurately and noninvasively detect ICAM1-positive cervical tumors, providing a simple and noninvasive approach to screen patients with high ICAM1 expression and
identify those who would benefit from ICAM1-ADC therapy. These MRI signal changes positively correlate with the level of tumoral antigen expression, which could be utilized to non-invasively
identify ICAM1+ patients who might benefit from our ADC therapy. DISCUSSION Given the complexity of the TME and its role in immunosuppression, patients with advanced or metastatic solid
tumors, including cervical cancer, often encounter significant challenges in treatment, especially due to the scarcity of targeted therapeutic options like ADCs [1, 8, 9]. To address this
challenge, our research endeavors to develop novel ADC therapeutics for reprogramming TME, with a specific focus on their effects on innate and adaptive immune cell dynamics. Concurrently,
we explore the synergy between ADC treatments and PD-1 immune checkpoint inhibitors, aiming to uncover novel therapeutic avenues. This study provides the inaugural experimental support for
ICAM1 as a viable ADC target in cervical cancer. By employing an unbiased and quantitative approach, we identified ICAM1 as a membrane protein that is overexpressed in cervical cancer cells
but not in adjacent normal cervical epithelia. Further analysis, including data from clinical specimens and the GEPIA database, indicated a correlation between ICAM1 overexpression and
adverse clinical outcomes in cervical cancer patients, underscoring its potential as a therapeutic target. Importantly, our findings regarding the endocytosis efficiency and antibody binding
to ICAM1-expressing cells underline the therapeutic relevance of ICAM1-targeted ADCs. Moreover, ICAM1’s involvement in TME dynamics and pathogenesis further validates its utility as a
target for ADC development [49]. Our investigation has yielded two promising ICAM1-targeted ADCs (I-DXd and I-MMAE) that have demonstrated preclinical efficacy and safety in models of
cervical cancer. Our comparative studies highlight the superior tumor-cell targeting and elimination capabilities of ICAM1-ADCs over conventional chemotherapy agents like Cis/Pac, in both in
vitro and in vivo settings. The design of these ADCs, incorporating enzyme-cleavable linkers (VC or GGFG) and potent cytotoxic agents (MMAE or DXd), achieves an optimal balance between
therapeutic efficacy and toxicity [50,51,52]. Notably, the GGFG-DXd linker used in I-DXd demonstrates advantages in the controlled release of payloads [53, 54], tumor penetration, and
bystander-killing effects [55]. I-DXd, in particular, possesses the dual capability of direct cancer cell ablation through antigen recognition and the ancillary eradication of protumor
VEGFA+ neutrophils (TANs), which contributes to a more immunologically favorable TME. Moreover, the successful development of I-DXd complements the limitations observed with the only other
ADC for cervical cancer, Tisotumab Vedotin, which targets tissue factor (TF) but is associated with significant ocular and peripheral neurotoxicity [56, 57]. I-DXd’s less cytotoxic payloads
suggest a reduced risk of these adverse effects. In addition, our current data indicate that most serum markers, including ALT, AST, ALP, CREA, UA, CHE, and TBIL, remained within the normal
range and did not show statistically significant differences compared to the PBS control group. Only a slight increase in UREA was observed, suggesting minimal renal impact under the current
dosing regimen. We acknowledge that extended treatment duration or additional dosing could increase the risk of cumulative toxicity. If treatment were prolonged, it would be essential to
monitor these markers over time to assess for any progressive trends. Increased frequency or duration of administration could potentially lead to elevated levels of UREA or other serum
markers, reflecting stress on renal or hepatic systems. Our current data, however, support that the single-agent ADC treatments remain within a safe range under the dosing schedule used.
Meanwhile, the potential of combining I-DXd with PD-1 inhibition warrants exploration in clinical settings, as preliminary evidence suggests this combination can enhance CD8 + T cell
recruitment, tumor suppression, and overall survival outcomes. Transcriptomic analysis post-treatment with I-DXd reveals significant modulation of the TME (Figs. 4 and 6), including
downregulation of neutrophil activation and upregulation of type I interferon signaling pathways, suggesting a comprehensive improvement in the immune landscape of treated tumors. These
findings, coupled with the observed efficacy of I-DXd in combination with PD-1 blockade, underscore the therapeutic potential of targeting ICAM1 in cervical cancer, particularly in TMEs
characterized by high mutational burdens and suppressed immune responses [58,59,60,61]. Furthermore, the most common adverse effect associated with ADC is peripheral neutropenia caused by
off-tumor toxicity of ADC [56, 57]. Our findings suggested that the use of granulocyte colony-stimulating factor (G-CSF) to manage ADC-induced neutropenia [57, 59], while standard, may
paradoxically enhance TAN infiltration and attenuate the antitumor efficacy of ADCs. Our data demonstrate that I-DXd selectively eliminates pro-tumor VEGFA+ TANs with high CSF3 expression
within the TME, mitigating the potential counterproductive effects of G-CSF and enhancing the immunomodulatory impact of ADC therapy. Through extensive preclinical evaluation, we provide a
mechanistic rationale for the combination of ADC and PD-1 inhibition in improving the TME and advancing the treatment of cervical cancer. In summary, we address the therapeutic challenge of
immunosuppression within the TME of advanced cervical cancer by the development of novel ADCs aimed at reprogramming the TME, with I-DXd showing particular promise due to its dual action on
cancer cell ablation and pro-tumor VEGFA + TAN eradication, fostering an immunologically favorable TME. The promising synergy of I-DXd with PD-1 inhibition, suggested by enhanced CD8 + T
cell recruitment and tumor suppression, alongside a comprehensive modulation of the TME, underscores the potential of ICAM1-targeted therapy to significantly advance cervical cancer
treatment. METHODS CELL LINES AND CULTURE Human cervical cancer cell lines (SiHa, CaSki, and ME180) were obtained from the cell bank of Zhejiang Cancer Hospital and Shanghai Cancer Hospital,
and normal cervical cells (HcerEpic) were purchased from Tongpai (Shanghai) Biotechnology Co., Ltd. Mouse colorectal cell line CT26 were purchased from China Center for Type Culture
Collection (Beijing, China). Normal human embryonic kidney HEK293T cell line was purchased from the American Type Culture Collection (Manassas, VA, USA). The culture medium MEM, RPMI-1640,
and fetal bovine serum were purchased from Gibco Company. All cells were cultured according to the recommended conditions. Medium containing 10% fetal bovine serum, and 1%
penicillin/streptomycin at 37 °C, and 5% CO2 incubator. The culture medium was changed once every 2–3 days, and the cells with a cell growth density of about 90% and in the logarithmic
growth phase were digested with trypsin for passage and follow-up experiments. ANTIBODY LIBRARIES AND REAGENTS PE anti-human CD9, Cat: 312106; PE anti-mouse/human CD15 (SSEA-1), Cat: 125606;
PE anti-human CD29, Cat: 303004; PE anti-human CD31, Cat: 303106; PE anti-human CD44, Cat: 397504; PE anti-human CD46, Cat: 352402; PE anti-human CD47, Cat: 323108; PE anti-human CD49a,
Cat: 328304; PE anti-human CD49b, Cat: 359308; PE anti-human CD49c (integrin α3), Cat: 343803; PE anti-human CD49e, Cat: 328010; PE anti-human CD50 (ICAM-3), Cat: 330005; PE anti-human
CD51/61, Cat:304406; PE anti-human CD56 (NCAM), Cat: 362508; PE anti-human CD62E, Cat: 336008; PE anti-human CD62P (P-Selectin), Cat: 304906; PE anti-human CD66a/c/e, Cat: 342304; PE
anti-human CD70, Cat: 355104;PE anti-human CD71, Cat: 334106; PE anti-human CD73 (Ecto-5’-nucleotidase), Cat: 344004; PE anti-human CD90 (Thy1), Cat:328110; PE anti-human CD105, Cat: 323206;
PE anti-human CD106, Cat: 305806;PE anti-human CD107a (LAMP-1), Cat: 328608; PE anti-human CD117 (c-kit), Cat: 375206; PE anti-human CD126 (IL-6Rα), Cat: 352804; PE anti-human CD140a
(PDGFRα), Cat: 323506; PE anti-human CD140b (PDGFRβ), Cat: 323606; PE anti-human CD141 (Thrombomodulin), Cat:344104; PE anti-human CD142, Cat: 365204; PE anti-human CD144 (VE-Cadherin), Cat:
348506; PE anti-human CD146, Cat: 361006; PE anti-human CD152 (CTLA-4), Cat: 349906; PE anti-human CD166, Cat: 343904; PE anti-human CD171 (L1CAM), Cat: 371604; PE anti-human CD181 (CXCR1)
Cat: 320608; PE anti-human CD184 (CXCR4) Cat: 306506; PE anti-human CD192 (CCR2) Cat:357206; PE anti-human CD195 (CCR5) Phospho (Ser349) Cat:321606; PE anti-human CD202b (Tie2/Tek)
Cat:334206; PE anti-human CD203c (E-NPP3) Cat:324606; PE anti-human CD221 (IGF-1R) Cat:351806; PE anti-human CD227 (MUC-1) Cat:355604; PE anti-human CD254 (TRANCE, RANKL) Cat:347504; PE
anti-human CD257 (BAFF, BLYS) Cat:366506; PE anti-human CD266 (Fn14, TWEAK Receptor) Cat:314004; PE anti-human CD271 (NGFR) Cat:345106; PE anti-human CD274 (B7-H1, PD-L1) Cat:329706; PE
anti-human CD276 (B7-H3) Cat:351004; PE anti-human CD304 (Neuropilin-1) Cat:354504; PE anti-human CD309 (VEGFR2) Cat:393004; PE anti-mouse CD317 (BST2, PDCA-1) Cat:127104; PE anti-human
CD324 (E-Cadherin) Cat:324106; PE anti-human CD325 (N-Cadherin) Cat:350805; PE anti-human CD326 (Ep-CAM) Cat:324206;PE anti-human CD340 (erbB2/HER-2) Cat:324406; PE anti-human CD371
(CLEC12A) Cat: 353604; PE anti-human EGFR Cat:352904; PE anti-human EphA2 Cat: 356804; PE anti-human ROR1 Cat: 357804; PE anti-human VEGFR-3 (FLT-4) Cat: 356204; PE anti-FOLR1 (Folate
Binding Protein) Cat: 908304; PE anti-human erbB3/HER-3 Cat:324706; PE anti-human Notch 3 Cat: 345406; PE anti-human PSMA (FOLH1) Cat: 342504; PE anti-human/mouse SSEA-3 Cat: 330312; PE
anti-human SSEA-4 Cat: 330406; PE anti-human SSEA-5 Cat: 355204; PE anti-human TM4SF Cat: 20367204; PE anti-human CD54 Cat: 353106; Antibody libraries and reagents are used to screen cancer
target candidates using FACS mothods. Other relative antibodies, PE mouse IgG1 (Cat: 400114), purified anti-human CD54 Antibody (Cat: 322702), and PE anti-mouse IgG1 Antibody (Cat: 406608)
were purchased from BioLegend. IHC METHODS Survival cervical cancer tissue chip (Cat: HUteS136Su01, Lot: XT17-039) was purchased from Shanghai Core Super Company. IHC staining was used to
detect the difference in ICAM1 target protein expression in 25 groups of human cervical cancer tissues, paracancerous tissues, and 86 cases of cervical tissues. The expression of ICAM1
(anti-ICAM1 antibody, ab1846317, Sigma; 1:1000) was analyzed semi-quantitatively by tissue microarray. The individual tissues were scored by a pathologist from Zhejiang Cancer Hospital, with
no knowledge of sample identity, based on both the intensity of staining and the proportion of positive cells within the section. The IHC staining scoring criteria of ICAM1 are defined as
shown in Fig. S2 and entailed a reference to the HER2 target and a differentiation of the positive areas with a tan depth (0, 1+, 2+, 3+). The method of ICAM1-H-Score scoring is detailed as
follows: the staining intensity (0, 1+, 2+, 3+) is multiplied by the percentage of positive cells in all tumor cells, and the resulting product represents the final score. The ICAM1-H-Score
score results are divided into two categories: ICAM1+ (Positive) and ICAM1-(negative). The ICAM1+ (positive) category is further subdivided into three expression levels: The ICAM1-H-Score
can be classified as low (1–10), moderate (10–40), or high (40–300). Conversely, ICAM1-(negative) is defined as negative (0), which indicates the complete absence of the specific ICAM1
expression. To illustrate, the staining intensity is 2+, the percentage of positive tumor cells is 15%, and the calculated ICAM1-H-Score is Moderate (30), which is determined as ICAM1 +
(Positive). Anti-Ly6G antibody (Cat: ab238132), anti-MPO antibody (Cat: ab208670), CD8α (D4W2Z) XP®Rabbit mab (Cat: 98941), anti-CD4 antibody (Cat: ab288724) are used for IHC staining in
animal models. FLOW CYTOMETRY FLOW CYTOMETRY Quantification of ICAM1 expression in cells: Flow cytometry (Beckman Coulter, CytoFLEX LX) was used to evaluate the expression of ICAM1 on the
surface of cervical cancer cells and normal cells. Quantitative analysis of cellular antigen expression was determined with Quantum TM Simply Cellular® microspheres (Bangs Laboratories) by
using a method provided by the manufacturer. One million cells were harvested from each cell line and rinsed twice with PBS. The harvested cells were blocked with 1% bovine serum albumin
(BSA) in PBS for 30 min at 4 °C. After BSA blocking, the cells were incubated with PE anti-human CD54 (Cat: 353106) for 1 h at 37 °C. PE anti-mouse IgG1 (Cat: 406608) was used as a control.
The cells were washed three times with 1% BSA in PBS, then resuspended in PBS, and the expression intensity of ICAM1 in each cell line was evaluated by flow cytometry. The PE anti-mouse IgG1
(Cat: 406608) and Purified anti-human CD54 (Cat: 322702) are purchased from BioLegend Company. Multi-colored flow cytometry: Fixable Viability Stain 780(Cat: 565388V500); Rat Anti-Mouse
CD45(30-F11)(Cat: 561487); FITC Rat Anti-CD11b(M1/70) (Cat: 557396); BV421 Hamster Anti-Mouse CD3e(145-2C11)(Cat: 562600); APC Rat Anti-Mouse CD4(RM4-5) (Cat: 553051); PE-Cy7 Rat Anti-Mouse
CD8a(53-6.7) (Cat: 552877); Purified Rat Anti-Mouse CD16/CD32 (Mouse BD Fc Block)(2.4G2) (Cat: 553141); Those antibodies Antibodies are used in multi-colored flow cytometry to detect changes
in the ratio and number of CD4/CD8 + T cells. CELL VIABILITY Viability assessment of drug killing: Three to five thousand cells were plated into 96-well plates and incubated with various
concentrations of drugs. After 72 h, CCK8 reagents were added and incubated for 1–4 h at 37 °C. Cell viability was examined by the CCK8 assay and detected absorption values at 450 nm.
Viability assessment of ICAM1 antibody: Ten thousand SiHa cells were plated into 96-well plates and incubated with purified anti-human CD54 antibody (Cat: 322702) at the indicated time
points (0-30-60-120-240 min). Cell viability was examined by the CCK8 assay and detected absorption values at 450 nm. DETERMINATION OF INTERNALIZATION The subcellular location of the target
protein was observed in different cell lines. One million cells were inoculated in confocal plates, and cultured overnight at 37 °C. The cells were washed in PBS and blocked in 1% BSA for 30
min at 4 °C, and incubated with fluorescent-labeled antibodies or drugs for 1 h. After staining with Hoechst, they were imaged with a confocal fluorescence microscope (NIKON, A1 HD25).
Cy3-NHS (Cat: R-H-6551), ICAM1-MMAE and ICAM1-MMAE are used to synthesize ICAM1-MMAE-cy3 and ICAM1-DXD-cy3. Mouse tumor infiltrating tissue neutrophil separation solution kit (Cat: P2430)
and mouse peripheral blood neutrophil separation kit (Cat: P9201) were used to isolate TAN and peripheral blood neutrophils. The confocal imaging was used to determine whether ICAM1-ADCs
could selectively enter cervical cancer cells and quickly transport them to intracellular lysosomes. The cells were inoculated, cultured overnight at 37 °C, and incubated with ICAM1 primary
antibody with 0 min, 30 min, 60 min, 120 min and 240 min at 37 °C. Then all cells were fixed with 4% paraformaldehyde, stained with Hoechst, and imaged with a confocal instrument. The
internalization efficiency was calculated by an established formula [62]: internalized% = (0 min MFI −t/min/MFI)/0 min × 100%. Purified anti-human CD54 antibody (Cat: 322702); PE-conjugated
rat anti-mouse IgG (BioLegend, San Diego, CA). Co-localization of ICAM1 antibody and lysosome were assessed by confocal fluorescent imaging. The cells were inoculated, incubated with ICAM1
primary antibody, fixation. Then permeabilization, and blocking were respectively performed by 4% formaldehyde, 0.5% TrotonX-100, and 0.5% BSA. The cells were incubated with LAMP1 antibody
at 4 °C overnight and then with cy5 anti-mouse lgG secondary Antibody for 2 h at room temperature. After staining Hoechst, confocal images were captured by a NIKON A1 HD25 microscope.
SYNTHESIS AND CHARACTERIZATION OF PREPARATION AND CHARACTERIZATION OF ADCS Monoclonal ICAM1 chimeric antibodies were covalently conjugated with two different linkers (MC-VC-PAB and MC-GGFG)
and two different warheads (MMAE and DXd) to create a group of ICAM1-ADCs, which includes ICAM1-MC-VC-PAB-MMAE (I-MMAE) and ICAM1-MC-GGFG-DXd (I-DXd). MMAE and DXd are commonly used warheads
in clinical ADCs. The DAR values of two types of ICAM1 ADCs were determined using hydrophobic interaction chromatography (HIC), ensuring their optimal efficacy and toxicity. IN VITRO
CYTOTOXICITY STUDIES Human cervical cancer cell lines (SiHa, CaSki) and normal cells (HcerEpic, 293T) were tested using the CCK8 kit (Cat: BS350B) from Bioshrp Company. The cells were
inoculated at a density of 3000 cells per well in 96-well petri dishes and cultured overnight. ICAM1-ADCs (I-MMAE, I-DXd), an ICAM1 monoclonal antibody, and two chemotherapy drugs (Cis,
Pac), were added to the wells at drug concentrations ranging from 0 μg/ml to 10 μg/ml. The addition was carried out with 10-fold continuous dilution, resulting in a total of eight
concentrations and three parallel wells per drug. After culturing the cells and drugs for 96 h, discard the original culture medium. Dilute the CCK8 detection reagent ten times with fresh
cell culture medium and add 100 PL/well to the 96-well plate. Incubate the plate at 37 °C for 1–4 h, and measure the absorbance (OD) at the absorption wavelength of 450 nm. ANIMAL
EXPERIMENTS All animal studies were performed according to the protocols approved by the Institutional Animal Care and Use Committee (IACUC) of Zhejiang Cancer Hospital. The in vivo
therapeutic efficacy of ICAM1-ADCs was evaluated in both standard and late-stage cervical tumor models. 4 × 106 SiHa cells suspended in 100 μl PBS (contained 50% Matrigel) were injected
subcutaneously into the right flank of 4-6-week-old female nude mice. When the tumor volume reached 50 to 80 mm3(standard model) or approximately 500 mm3 (late-stage model), the mice were
randomized into 5 groups (_n_ = 6 or 7 per group) and dealt with different treatments of PBS, Cis, ICAM1 monoclonal antibody (I Ab), I-DXd or I-MMAE in an equivalent dosage of 5mg/kg with QW
× 2 manner via tail vein injection. In addition, for the combination therapy study, 6 × 106 CT26-hICAM1 cells suspended in 100 μL PBS (contained 50% Matrigel) were injected subcutaneously
into the right flank of 4–6-week-old female BALB/c mice. When the tumor volume reached 300 mm3, BALB/c mice were randomized into 4 groups of 7 and treated with PBS, 10 mg/kg anti-mouse PD-1
antibody (InVivoPlus, Cat: BP0146) for Q3D × 2, 5 mg/kg I-DXd for Q3D × 2, and combination with I-DXd and PD-1. Tumor sizes and body weight were recorded every other day or daily. The long
diameter (a) and short diameter (b) of tumor nodules were measured with calipers and the tumor volume was calculated according to the tumor volume: V = a×b2/2. The tumor-bearing mice were
recorded and euthanized at the end of the study, or when tumors reached 1500 mm3. At the end of the experiments, blood serum was collected from each experimental group (_n_ = 2–5 per group).
The serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), total bilirubin (TB), blood concentration of creatinine (BUN) and
creatinine (Cre) and other liver, kidney and inflammation indicators were determined with biochemical analyzer Hitachi 7180 (Hitachi, Yokohama). WESTERN BLOT ANALYSIS Isolation of TANs and
peripheral blood neutrophils: Mouse tumors infiltrating tissue neutrophil separation solution kit (Cat: P2430) and mouse peripheral blood neutrophil separation kit (Cat: P9201) were used to
isolate TANs and peripheral blood neutrophils. Peripheral blood and tumor tissues were collected from three tumor-bearing female nude mice at the same time. The tumor tissue was minced and
ground into a cell suspension and then filtered through a 70 μm filter membrane to form a single cell suspension. According to the manufacturer's instructions, the target cells were
isolated and collected for further use. TANs, peripheral blood neutrophils, and mouse cervical cancer cells (U14-ICAM1) were lysed by SDS lysis buffer supplemented with Phosphatase Inhibitor
Cocktail. All samples were analyzed by SDS-PAGE and then transferred to PVDF membranes. The membranes were blocked with 5% bovine serum albumin (Cat: D8340) for 1 h at room temperature and
then incubated with the primary antibody anti-Cathepsin B (Cat: ab214428) while anti-β-Actin (Cat: EM21002) as control at 4°C overnight. Then the membranes were incubated with secondary
antibodies for 1h at room temperature. The protein bands were detected using the ECL reagent (Cat: BL523B) and Amersham ImageQuant 800 systems. RNA-SEQ ANALYSIS RNA extraction, cDNA library
preparation, RNA sequencing, quality control, and transcriptome profiling of endpoint tumors in different treatment groups (PBS, ICAM1 mAb, I-DXd, and I-MMAE, three repeated samples per
group) of late-stage tumor mouse models were performed by Novogene (Beijing, China). The GO enrichment analysis and Reactome pathway analysis enrichment of DEGs were conducted between PBS,
Cis, I-DXd and I-MMAE treatment groups. The clinical relevance and prognosis of VEGFA+ TANs in the GSE63514 database and TCGA cohort were assessed in gene set variation analysis (GSVA). In
addition, The CIBERSORT and xCELL algorithms were employed to deconvolute bulk RNA-seq profiles and quantify the infiltration of fibroblasts, immune cells (such as CD8+、CD4+ T cells), and
other cell types. The changes between high and low TAN-related GSVA score groups in the proportion of immune cells in the TME of cancer patients in the TCGA cohort were assessed in EPIC
(https://gfellerlab.shinyapps.io/EPIC_1-1). I-GD PREPARATION AND MRI IMAGING I-Gd, lgG-Gd drugs prepared: The DTPAA is gradually added to the lgG or ICAM1 antibody, and the reaction is
continuously monitored in the two buffers, resulting in the production of lgG-Gd and I-Gd [47]. Briefly, DTPAA (1 mg) was slowly added to lgG or ICAM1 Ab (1 mg) in 1 ml of buffer system 1
(NaHCO3 buffer, pH = 9.0), and the solution should be kept in the dark at 37 °C for four hours. The DTPA-conjugated IgG or ICAM1 Ab was transferred to a dialysis bag and placed in a beaker
of ultra-pure water for dialysis overnight, with unbound small molecules being filtered out. Subsequently, the liquid within the dialysis bag was subjected to freeze-drying at a temperature
of −80 °C, resulting in the formation of a powder. This powder was then dissolved in 1 ml of buffer 2 (sodium citrate buffer, pH = 6.5), and subsequently transferred to a tube containing 0.3
mg of Gadolinium (III) chloride hexahydrate (Cat: G7532-5g) for 24 h rotated at 37 °C. The free Gd3+ was removed by Ultra-4 Centrifugal Filter (30 K MWCO), and IgG-Gd or I-Gd was
redispersed in PBS and measured the concentration of BCA protein for subsequent use. MRI imaging: 6-week-old female tumor-bearing (SiHa) nude mice were randomized into two groups (I-Gd and
lgG-Gd, n = 4 or 5 per group), when the tumor volume reached 500 to 1000 mm3.The target group (I-Gd) and the control group (lgG-Gd) were both administered an equivalent dose of 5 mg/kg via
the tail vein, and MRI imaging was performed 24 hours later. The MRI instrument (Bruker BioSpin MRI, 7.0-T) was used to perform T1 and T2-weighted MR imaging to quantify MRI signal changes.
MRI gradient echo sequence was given to each group. The following parameters were used: TR/TE = 1300/7.2 ms, 1 mm slice thickness, FOV 71 × 85 mm, and FOV 256 × 256 matrix. QUANTIFICATION
AND STATISTICAL ANALYSIS GraphPad Prism 8.0 and FlowJo V10 were used to analyze the experimental data, and the error bars indicate SEM. Statistical significance was calculated by one-way
ANOVA with Tukey’s multiple comparison test. The difference between the two groups was analyzed by _t_-test. Kaplan–Meier survival using the log-rank (Mantel–Cox) test for the clinical
survival data or the Gehan-Wilcoxon test for the mouse survival data, followed by Bonferroni’s correction for multiple comparisons. *_P_ < 0.05, **_P_ < 0.01, ***_P_ < 0.001,
****_P_ < 0.0001, ns, not significant. GraphPad Prism 8.0 is used to draw related pictures of the experimental data. Quantify the signal intensity of the tumor and draw the region of
interest (ROI) around the whole tumor at the same imaging depth. The pixel intensity is calculated by ImageJ software and normalized to ROI. DATA AVAILABILITY All data in our study are
available upon reasonable request. REFERENCES * Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and
mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin, 2018. 2021;71:209–249. Article Google Scholar * Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA
Cancer J Clin 2023;73:17–48. Article PubMed Google Scholar * Fiorica JV. The role of topotecan in the treatment of advanced cervical cancer. Gynecol Oncol 2003;90:S16–S21. Article CAS
PubMed Google Scholar * McAlpine JN, Temkin SM, Mackay HJ. Endometrial cancer: not your grandmother’s cancer. Cancer 2016;122:2787–2798. Article PubMed Google Scholar * Hu K, Wang W,
Liu X, Meng Q, Zhang F. Comparison of treatment outcomes between squamous cell carcinoma and adenocarcinoma of cervix after definitive radiotherapy or concurrent chemoradiotherapy. Radiat
Oncol 2018;13:249. Article CAS PubMed PubMed Central Google Scholar * Olawaiye AB, Baker TP, Washington MK, Mutch DG. The new (Version 9) American Joint Committee on Cancer tumor, node,
metastasis staging for cervical cancer. CA Cancer J Clin 2021;71:287–298. Article PubMed Google Scholar * Zheng RS, Sun KX, Zhang SW, Zeng HM, Zou XN, Chen R, et al. [Report of cancer
epidemiology in China, 2015]. Zhonghua Zhong Liu Za Zhi 2019;41:19–28. CAS PubMed Google Scholar * Vu M, Yu J, Awolude OA, Chuang L. Cervical cancer worldwide. Curr Probl Cancer.
2018;42:457–465. Article PubMed Google Scholar * Kondrashov A, Sapkota S, Sharma A, Riano I, Kurzrock R, Adashek JJ. Antibody-drug conjugates in solid tumor oncology: an effectiveness
payday with a targeted payload. Pharmaceutics 2023;15:2160. Article CAS PubMed PubMed Central Google Scholar * Wu Q, Qian W, Sun X, Jiang S. Small-molecule inhibitors, immune checkpoint
inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol 2022;15:143. Article CAS PubMed PubMed Central Google Scholar *
Hainsworth JD, Meric-Bernstam F, Swanton C, Hurwitz H, Spigel DR, Sweeney C, et al. Targeted therapy for advanced solid tumors on the basis of molecular profiles: results from my pathway, an
open-label, phase iia multiple basket study. J Clin Oncol 2018;36:536–542. Article CAS PubMed Google Scholar * Coleman RL, Lorusso D, Gennigens C, González-Martín A, Randall L, Cibula
D, et al. Efficacy and safety of tisotumab vedotin in previously treated recurrent or metastatic cervical cancer (innova TV 204/GOG-3023/ENGOT-cx6): a multicentre, open-label, single-arm,
phase 2 study. Lancet Oncol 2021;22:609–619. Article CAS PubMed Google Scholar * Shitara K, Bang YJ, Iwasa S, Sugimoto N, Ryu MH, Sakai D, et al. Trastuzumab deruxtecan in previously
treated HER2-positive gastric cancer. N Engl J Med 2020;382:2419–2430. Article CAS PubMed Google Scholar * Breij EC, de Goeij BE, Verploegen S, Schuurhuis DH, Amirkhosravi A, Francis J,
et al. An antibody-drug conjugate that targets tissue factor exhibits potent therapeutic activity against a broad range of solid tumors. Cancer Res. 2014;74:1214–1226. Article CAS PubMed
Google Scholar * Wei Q, Yang T, Zhu J, Zhang Z, Yang L, Zhang Y, et al. Spatiotemporal quantification of HER2-targeting antibody-drug conjugate bystander activity and enhancement of solid
tumor penetration. Clin Cancer Res 2024;30:984–997. Article CAS PubMed Google Scholar * Xue R, Zhang Q, Cao Q, Kong R, Xiang X, Liu H, et al. Liver tumour immune microenvironment
subtypes and neutrophil heterogeneity. Nature 2022;612:141–147. Article CAS PubMed Google Scholar * Zhou J, Nefedova Y, Lei A, Gabrilovich D. Neutrophils and PMN-MDSC: their biological
role and interaction with stromal cells. Semin Immunol 2018;35:19–28. Article CAS PubMed Google Scholar * Li MO, Wolf N, Raulet DH, Akkari L, Pittet MJ, Rodriguez PC, et al. Innate
immune cells in the tumor microenvironment. Cancer Cell 2021;39:725–729. Article CAS PubMed Google Scholar * Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z, et al. Current perspectives
on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol Cancer 2019;18:130. Article PubMed PubMed Central Google Scholar * Guo P,
Huang J, Wang L, Jia D, Yang J, Dillon DA, et al. ICAM-1 as a molecular target for triple negative breast cancer. Proc Natl Acad Sci USA. 2014;111:14710–14715. Article CAS PubMed PubMed
Central Google Scholar * Doronina SO, Toki BE, Torgov MY, Mendelsohn BA, Cerveny CG, Chace DF, et al. Development of potent monoclonal antibody auristatin conjugates for cancer therapy.
Nat Biotechnol 2003;21:778–784. Article CAS PubMed Google Scholar * Shen BQ, Xu K, Liu L, Raab H, Bhakta S, Kenrick M, et al. Conjugation site modulates the in vivo stability and
therapeutic activity of antibody-drug conjugates. Nat Biotechnol 2012;30:184–189. Article CAS PubMed Google Scholar * Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, et
al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising antitumor efficacy with differentiation from T-DM1. Clin Cancer Res.
2016;22:5097–5108. Article CAS PubMed Google Scholar * Gedik ME, Saatci O, Oberholtzer N, Uner M, Akbulut Caliskan O, Cetin M, et al. Targeting TACC3 induces immunogenic cell death and
enhances T-DM1 response in HER2-positive breast cancer. Cancer Res 2024;84:1475–1490. Article CAS PubMed PubMed Central Google Scholar * Xu Y, Lv J, Liu F, Wang J, Liu Y, Kong C, et al.
Tumor microenvironment remodeling-mediated sequential drug delivery potentiates treatment efficacy. Adv Mater 2024;36:e2312493. Article PubMed Google Scholar * Yu Y, Jiang L, Wang H,
Shen Z, Cheng Q, Zhang P, et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood 2020;136:726–739. Article CAS PubMed PubMed Central Google
Scholar * Boehm O, Zur B, Koch A, Tran N, Freyenhagen R, Hartmann M, et al. Clinical chemistry reference database for Wistar rats and C57/BL6 mice. Biol Chem 2007;388:547–554. Article CAS
PubMed Google Scholar * Valadez-Cosmes P, Raftopoulou S, Mihalic ZN, Marsche G, Kargl J. Myeloperoxidase: growing importance in cancer pathogenesis and potential drug target. Pharmacol
Ther 2022;236:108052. Article CAS PubMed Google Scholar * Rymaszewski TE, Yimbesalu JP, Gelman AE, Jarzembowski JA, Zhang H, Pritchard KA Jr, Vikis HG, The role of et al. The role of
neutrophil myeloperoxidase in models of lung tumor development. Cancers 2014;6:1111–1127. Article PubMed PubMed Central Google Scholar * Ng MSF, Kwok I, Tan L, Shi C, Cerezo-Wallis D,
Tan Y, et al. Deterministic reprogramming of neutrophils within tumors. Science 2024;383:6493. Article Google Scholar * Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani
A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer 2020;20:485–503. Article CAS PubMed Google Scholar * Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z,
Chen EB, et al. Tumor-associated neutrophils recruit macrophages and T-regulatory cells to promote progression of hepatocellular carcinoma and resistance to sorafenib. Gastroenterology
2016;150:1646–1658. Article CAS PubMed Google Scholar * Wu Y, Ma J, Yang X, Nan F, Zhang T, Ji S, et al. Neutrophil profiling illuminates anti-tumor antigen-presenting potency. Cell
2024;187:1422–1439. Article CAS PubMed Google Scholar * Zhao Y, Liu Z, Liu G, Zhang Y, Liu S, Gan D, et al. Neutrophils resist ferroptosis and promote breast cancer metastasis through
aconitate decarboxylase 1. Cell Metab 2023;35:1688–1703. Article CAS PubMed PubMed Central Google Scholar * Chen X, Gula H, Pius T, Ou C, Gomozkova M, Wang LX, et al. Immunoglobulin G
subclasses confer protection against Staphylococcus aureus bloodstream dissemination through distinct mechanisms in mouse models. Proc Natl Acad Sci USA 2023;120:2220765120. Article Google
Scholar * Jönsson F, de Chaisemartin L, Granger V, Gouel-Chéron A, Gillis CM, Zhu Q, et al. An IgG-induced neutrophil activation pathway contributes to human drug-induced anaphylaxis. Sci
Transl Med 2019;11:eaat1479. Article PubMed Google Scholar * Mysore V, Cullere X, Mears J, Rosetti F, Okubo K, Liew PX, et al. FcγR engagement reprograms neutrophils into antigen
cross-presenting cells that elicit acquired anti-tumor immunity. Nat Commun 2021;12:4791. Article CAS PubMed PubMed Central Google Scholar * Karin E, de Visser KE, Joyce JA. The
evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell 2023;41:374–403. Article Google Scholar * Lukhele S, Rabbo DA, Guo M, Shen J, Elsaesser HJ,
Quevedo R, et al. The transcription factor IRF2 drives interferon-mediated CD8+ T cell exhaustion to restrict anti-tumor immunity. Immunity 2022;55:2369–2385. Article CAS PubMed PubMed
Central Google Scholar * Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol 2003;3:133–146. Article CAS PubMed Google Scholar *
Alimena S, Lykken JM, Tiro JA, Chubak J, Kamineni A, Haas JS, et al. Timing of colposcopy and risk of cervical cancer. Obstet Gynecol 2023;142:1125–1134. CAS PubMed PubMed Central Google
Scholar * Wang S, Wang J, Liu C, Yang L, Tan X, Chen S, et al. Neoplastic ICAM-1 protects lung carcinoma from apoptosis through ligation of fibrinogen. Cell Death Dis 2024;15:605. Article
CAS PubMed PubMed Central Google Scholar * Lim EJ, Kang JH, Kim YJ, Kim S, Lee SJ. ICAM-1 promotes cancer progression by regulating SRC activity as an adapter protein in colorectal
cancer. Cell Death Dis 2022;13:968. Article PubMed PubMed Central Google Scholar * Cho BJ, Choi YJ, Lee MJ, Kim JH, Son GH, Park SH, et al. Classification of cervical neoplasms on
colposcopic photography using deep learning. Sci Rep. 2020;10:13652. Article CAS PubMed PubMed Central Google Scholar * Tan JH, Wrede CD. New technologies and advances in colposcopic
assessment. Best Pract Res Clin Obstet Gynaecol 2011;25:667–677. Article PubMed Google Scholar * Bui TM, Wiesolek HL, Sumagin R. ICAM-1: a master regulator of cellular responses in
inflammation, injury resolution, and tumorigenesis. J Leukoc Biol 2020;108:787–799. Article CAS PubMed Google Scholar * Huang J, Agoston AT, Guo P, Moses MA. A rationally designed ICAM1
antibody drug conjugate for pancreatic cancer. Adv Sci 2020;7:2002852. Article CAS Google Scholar * Guo P, Huang J, Zhu B, Huang AC, Jiang L, Fang J, et al. A rationally designed ICAM1
antibody drug conjugate eradicates late-stage and refractory triple-negative breast tumors in vivo. Sci Adv 2023;9:7866. Article Google Scholar * Bai R, Pettit GR, Hamel E. Dolastatin 10,
a powerful cytostatic peptide derived from a marine animal. Inhibition tubulin polymerization mediated vinca alkaloid binding domain. Biochem Pharmacol 1990;39:1941–1949. Article CAS
PubMed Google Scholar * Nakada T, Masuda T, Naito H, Yoshida M, Ashida S, Morita K, et al. Novel antibody drug conjugates containing exatecan derivative-based cytotoxic payloads. Bioorg
Med Chem Lett 2016;26:1542–1545. Article CAS PubMed Google Scholar * Sochaj AM, Świderska KW, Otlewski J. Current methods for the synthesis of homogeneous antibody-drug conjugates.
Biotechnol Adv 2015;33:775–784. Article CAS PubMed Google Scholar * Li BT, Michelini F, Misale S, Cocco E, Baldino L, Cai Y, et al. HER2-mediated internalization of cytotoxic agents in
ERBB2 amplified or mutant lung cancers. Cancer Discov 2020;10:674–687. Article CAS PubMed PubMed Central Google Scholar * Modi S, Park H, Murthy RK, Iwata H, Tamura K, Tsurutani J, et
al. Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-low-expressing advanced breast cancer: results from a phase Ib study. J Clin Oncol 2020;38:1887–1896.
Article CAS PubMed PubMed Central Google Scholar * Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor
receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci 2016;107:1039–1046. Article CAS PubMed PubMed Central Google
Scholar * O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol 2019;16:151–167. Article PubMed Google Scholar *
Yamaguchi K, Bang YJ, Iwasa S, Sugimoto N, Ryu MH, Sakai D, et al. Trastuzumab deruxtecan in anti-human epidermal growth factor receptor 2 treatment-naive patients with human epidermal
growth factor receptor 2-low gastric or gastroesophageal junction adenocarcinoma: exploratory cohort results in a phase II trial. J Clin Oncol 2023;41:816–825. Article CAS PubMed Google
Scholar * Terui Y, Rai S, Izutsu K, Yamaguchi M, Takizawa J, Kuroda J, et al. A phase 2 study of polatuzumab vedotin + bendamustine + rituximab in relapsed/refractory diffuse large B-cell
lymphoma. Cancer Sci 2021;112:2845–2854. Article CAS PubMed PubMed Central Google Scholar * Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, et al. Trastuzumab emtansine for residual
invasive HER2-positive breast cancer. N Engl J Med 2019;380:617–628. Article PubMed Google Scholar * Breast Cancer Group, Branch of Oncologist, Chinese Medical Doctor Association;
International Medical Exchange Society, Chinese Anti-Cancer Association. [Chinese expert consensus of antibody-drug conjugate toxicity management for breast cancer]. Zhonghua Zhong Liu Za
Zhi 2022;44:913–927. * Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous and multifaceted. Nat Rev Immunol 2022;22:173–187. Article CAS PubMed Google Scholar * Gungabeesoon J,
Gort-Freitas NA, Kiss M, Bolli E, Messemaker M, Siwicki M, et al. A neutrophil response linked to tumor control in immunotherapy. Cell. 2023;186:1448–1464. Article CAS PubMed PubMed
Central Google Scholar * Liao-Chan S, Daine-Matsuoka B, Heald N, Wong T, Lin T, Cai AG, et al. Quantitative assessment of antibody internalization with novel monoclonal antibodies against
Alexa fluorophores. PLoS One 2015;10:e0124708. Article PubMed PubMed Central Google Scholar Download references ACKNOWLEDGEMENTS This study was supported by the National Key R&D
Program of China (2023YFC3404003), the National Natural Science Foundation of China (NSFC; 82172598, 82373782), the Key Research and Development Program of Zhejiang Province (2024SDYXS0001),
the Natural Science Foundation of Zhejiang Province, China (LZ22H310001), the 551 Health Talent Training Project of Health Commission of Zhejiang Province, China, the Agricultural and
Social Development Research Project of Hangzhou Municipal Science and Technology Bureau (2022ZDSJ0474), Zhejiang Leading Innovation and Entrepreneurship Team (No. 2022R01006), Qiantang
Interdisciplinary Research Grant, and the Zhejiang Provincial Public Welfare Technology Applied Research Program (LTGY23H160013 and LGF21H160008). The authors acknowledge the use of
instruments at the Shared Instrumentation Core Facility at the Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou. AUTHOR INFORMATION Author notes * These authors
contributed equally: Huarong Tang, Peng Guo, Tao Zhu. AUTHORS AND AFFILIATIONS * School of Materials Science and Engineering, Faculty of Medicine, Tianjin University, Tianjin, China Shili
Yao & Peng Guo * Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China Shili Yao, Lu Sun, Ye Lu, Rui Xu, Tong
Yang, Huarong Tang, Peng Guo & Tao Zhu * Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China Lu Sun, Huarong Tang, Peng Guo & Tao Zhu * Department of
Pathology, Zhejiang Cancer Hospital, Hangzhou, China Xiu Zhu & Tao Zhu * Institute of Molecular Medicine, Hangzhou Institute for Advanced Study (UCAS), Hangzhou, China Rui Xu *
Department of Gynecological Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China Huarong Tang Authors * Shili Yao View author publications You can also search for this author inPubMed
Google Scholar * Lu Sun View author publications You can also search for this author inPubMed Google Scholar * Ye Lu View author publications You can also search for this author inPubMed
Google Scholar * Xiu Zhu View author publications You can also search for this author inPubMed Google Scholar * Rui Xu View author publications You can also search for this author inPubMed
Google Scholar * Tong Yang View author publications You can also search for this author inPubMed Google Scholar * Huarong Tang View author publications You can also search for this author
inPubMed Google Scholar * Peng Guo View author publications You can also search for this author inPubMed Google Scholar * Tao Zhu View author publications You can also search for this author
inPubMed Google Scholar CONTRIBUTIONS Shili Yao: conceptualization, formal analysis, funding acquisition, validation, investigation, visualization, methodology, writing—original draft, and
project administration. Lu Sun: resources, data curation, writing—review and editing. Ye Lu: software and formal analysis. Xiu Zhu: data curation, formal analysis, and investigation. Rui Xu:
validation, investigation, and visualization. Tong Yang: data curation and formal analysis. Huarong Tang: Supervision and project administration. Peng Guo: conceptualization, supervision,
project administration, and writing—review and editing. Tao Zhu: resources, supervision, and project administration. CORRESPONDING AUTHORS Correspondence to Huarong Tang, Peng Guo or Tao
Zhu. ETHICS DECLARATIONS COMPETING INTERESTS Shili Yao, Peng Guo, Huarong Tang, and Tao Zhu are co-inventors of a patent application filed (202210334398.7) by Zhejiang Cancer Hospital. The
other authors declare no competing interests. ETHICS APPROVAL AND CONSENT TO PARTICIPATE All animal experiments were approved by the Institutional Animal Care and Use Committee (IACUC) of
Zhejiang Cancer Hospital (Zhejiang, China). CONSENT FOR PUBLICATION All authors gave their consent to the publication. ADDITIONAL INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral
with regard to jurisdictional claims in published maps and institutional affiliations. Edited by Hans-Uwe Simon SUPPLEMENTARY INFORMATION SUPPLEMENTARY FIGURES 1–9 AND CAPTIONS RIGHTS AND
PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any
medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The
images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not
included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. Reprints and permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Yao, S., Sun, L.,
Lu, Y. _et al._ Eliminating VEGFA+ tumor-associated neutrophils by antibody-drug conjugates boosts antitumor immunity and potentiates PD-1 immunotherapy in preclinical models of cervical
cancer. _Cell Death Dis_ 16, 115 (2025). https://doi.org/10.1038/s41419-025-07402-9 Download citation * Received: 28 August 2024 * Revised: 08 January 2025 * Accepted: 27 January 2025 *
Published: 19 February 2025 * DOI: https://doi.org/10.1038/s41419-025-07402-9 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable
link Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative