You are on page 1of 16

Biosensors and Bioelectronics 240 (2023) 115644

Contents lists available at ScienceDirect

Biosensors and Bioelectronics


journal homepage: www.elsevier.com/locate/bios

Current progress, strategy, and prospects of PD-1/PDL-1 immune


checkpoint biosensing platforms for cancer diagnostics, therapy
monitoring, and drug screening
Katarzyna Ratajczak 1, Hubert Grel 1, Piotr Olejnik 1, Slawomir Jakiela **, Magdalena Stobiecka *
Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland

A R T I C L E I N F O A B S T R A C T

Keywords: Recent technological advancements in testing and monitoring instrumentation have greatly contributed to the
Optical biosensors progress in cancer treatment by surgical, chemotherapeutic and radiotherapeutic interventions. However, the
Surface plasmon resonance mortality rate still remains high, calling for the development of new treatment strategies with higher efficacy.
Microfluidic systems
Extensive efforts driven in this direction have included broadening of early cancer screening and applying
PD-1/PD-L1 pathway
Immune checkpoint inhibitors screening
innovative theranostic nanotechnologies. They have been supported by platforms introduced to enable the
Exosomal PD-L1 detection and monitoring of cancer biomarkers, inhibitors, and other agents, able to slow down cancer pro­
gression and prevent metastasis. Despite of the well-recognized principles of the immune checkpoint blockade,
the efficacy of immunotherapy achieved so far does not meet the well-founded expectations. For a successful
cancer treatment, highly sensitive, robust, and inexpensive multiplex biosensors have to be designed to aid in the
biomarkers monitoring and in the development of new inhibitors. In this review, we provide an overview of the
efforts undertaken to aid in the development and monitoring of anticancer immunotherapy, based on the pro­
grammed cell-death immune checkpoint (PD-1/PDL-1) blockade, by designing biosensors for the detection of
relevant cancer biomarkers and their inhibitors screening. This review also emphasizes alternative targets made
by exosomes carrying PD-L1 overexpressed in cancer cells and passed into the excreted exosomes. Evaluated are
also novel targeted drug delivery nanocarriers, providing simultaneous biosensing, thereby contributing to the
emerging immune checkpoint cancer therapy. On the basis of the current trends and the emerging technologies,
future perspectives of cancer diagnostics and treatment monitoring using biosensing platforms are projected.

1. Introduction well recognized immune checkpoint mechanisms (Dong et al., 1999;


Garrett and Collins, 2011; Pardoll, 2012). Since the attempts at the
Considerable progress has recently been achieved in cancer treat­ application of cancer immunotherapy have been met with varying de­
ment, with advancements in precision surgical techniques, nanotech­ gree of success, the great potential of immune checkpoint controlling
nology driven targeted chemotherapy, and multi-modal radiotherapy. has yet to be realized (Brahmer et al., 2012; Goodman et al., 2017; Ribas,
These advancements have strongly been supported by novel diagnostics 2012). Due to the projections of the continuing growth in the number of
and monitoring technologies enabling largely successful management of diagnosed cancer cases (2020; Siegel et al., 2021) (Fig. 1A), extensive
cancer growth retardation and prevention of metastasis (Corthay, 2014; efforts of scientists and clinicians have been focused on broadening of
Couzin-Frankel, 2013; Farkona et al., 2016; Janssen et al., 2017; Kauf­ early cancer screening and on the development of new therapeutic ap­
man et al., 2019; Weigmann, 2016). However, the use of less invasive proaches, including, among others, the induced immunotherapies
treatments involving the organism’s own defenses against cancer, based against cancer. In this review, we focus on cancer immunotherapies,
on the immune system ability to selectively kill cancer cells, has only with particular attention paid to the progress in the development of
been at the beginning stages of development, despite of the generally methods and biosensing devices for monitoring of biomarkers involved

* Corresponding author.
** Corresponding author.
E-mail addresses: slawomir_jakiela@sggw.edu.pl (S. Jakiela), magdalena_stobiecka@sggw.edu.pl (M. Stobiecka).
1
Katarzyna Ratajczak, Hubert Grel and Piotr Olejnik contributed equally to this work.

https://doi.org/10.1016/j.bios.2023.115644
Received 25 July 2023; Received in revised form 22 August 2023; Accepted 26 August 2023
Available online 28 August 2023
0956-5663/© 2023 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

in the immune checkpoint controlling, such as the programmed cell A low-level expression of PD-L1 has been encountered in many cell
death protein 1 (PD-1), programmed cell death ligand 1 (PD-L1), in­ types. However, an elevated expression of PD-L1 on the surface of tumor
hibitors, and moderators of immune checkpoints, as well as the drug cells has generally been observed and associated with worse prognosis
molecules under development. Hence, we aim in this review at evalu­ for many tumors (Mahoney et al., 2015). PD-L1 causes the inhibition of
ating novel platforms for monitoring of biomarkers and immunodrugs, kinase signaling pathways upon binding to the immune cell PD-1 re­
such as the biosensors arrays, microfluidic devices, optical fiber grat­ ceptors, resulting in suppressing of the T cells activity and taming of
ings, functionalized gold nanoparticles and nano-shells, gated resonance immune response (Aguiar et al., 2017; Davis and Patel, 2019; Incorvaia
energy transfer (gRET) plasmonic nanoprobes and other biosensing et al., 2019; Patel and Kurzrock, 2015). In this way, cancer cells
devices. expressing PD-L1 can easily evade immune reaction of the host and grow
Among various modalities of cancer treatment, the immunotherapy freely by binding T cells (Santarpia and Karachaliou, 2015).
is by far the most natural one as it utilizes the components of the or­ The crystal structure of human PD-1/human PD-L1 complex was
ganism’s own immune system to fight and eradicate cancer cells (Asa­ recently reported by Zak et al., (2015). The Authors have shown mo­
dujjaman et al., 2020; Couzin-Frankel, 2013; Farkona et al., 2016; lecular details of the human PD-1/PD-L1 interaction based on the X-ray
Kaufman et al., 2019; Xia et al., 2019). Immunotherapy complements structure of the complex. They have identified two spots, among three
commonly applied but highly invasive cancer treatments, including major active spots, which may facilitate the formation of a druggable
surgery, radiotherapy, and conventional chemotherapy. Due to the key pharmacophore capable of disrupting the PD-1/PD-L1 interaction.
features of immune-mediated therapy, which include biocompatibility, The mechanisms of the immune activity control have been eluci­
specificity, breadth of response, and memory (Kaufman et al., 2019), dated upon the discovery of immune checkpoints, leading to the
enabling noninvasive cancer treatment with no collateral damage to development of anticancer immunotherapy concept, recognized in 2018
healthy cells, it is becoming a powerful clinical strategy against cancer. with a Nobel Prize in Physiology or Medicine, awarded to James Allison
The group of immunotherapies consists of five distinct classes that and Tasuku Honjo (Huang and Chang, 2019; Zang, 2018). Honjo and his
include: (i) immunomodulators; (ii) cell-based immunotherapies (e.g. group discovered, on T cells, the programmed death molecule-1 (PD-1),
chimeric antigen receptor T cells (CAR T)); (iii) antibody-based targeted a member of the immunoglobulin superfamily (Ishida et al., 1992),
therapies that co-stimulate cells or block the checkpoint pathways (e.g. while Allison and his colleagues have shown that the monoclonal anti­
PD-1/PD-L1); (iv) cancer vaccines; and (v) oncolytic viruses, designed to body against the immunosuppressive molecule, cytotoxic T-lymphocy­
selectively infect and kill cancer cells, while inducing systemic te-associated protein 4 (CTLA-4), blocked the CTLA-4 function and
anti-tumor immune response and memory (García-Aranda and enhanced the anti-tumor immunity. The mice treated with anti-CTLA-4
Redondo, 2019; Gong et al., 2019; Iwai et al., 2002; Marshall and antibodies completely rejected their tumors (Leach et al., 1996).
Djamgoz, 2018; Ribas, 2012; Xie et al., 2018) (Fig. 1B). The excellent recent reviews related to the PD-1/PD-L1 cancer
The immune system is able to recognize most of the cancer cells immunotherapy have mainly focused on the mechanisms involving
(Corthay, 2014; Gonzalez et al., 2018; Janssen et al., 2017; Pandya et al., PD1/PD-L1 checkpoint inhibitors and their applications in tumor
2016). Unfortunately, some cancer cells can evade the immune system immunotherapy (Doroshow et al., 2021; Liu et al., 2021), the develop­
and grow (Marshall and Djamgoz, 2018). It has been found that the ment of biophysical and biochemical assays for binding affinity mea­
prevailing mechanism of immune system evasion involves activation of surements to identify small molecule inhibitors against PD-1/PD-L1 (Liu
the checkpoints of the programmed cell death (PD) pathway (Han et al., et al., 2021), as well as the meta-analysis of predictive biomarkers for
2020). immune checkpoint inhibitor (ICI) therapy (Mariam et al., 2023). In this
PD-1, also known as PDCD1 or CD279 (cluster of differentiation No. review, we focus on the evaluation of the progress in the development of
279), is highly expressed in the membrane of activated T cells, B cells, two classes of monoclonal antibodies (anti-PD-1 and anti-PD-L1) and the
and natural killer cells and its main role is to regulate the immune aptamers-based biosensors for the detection of the programmed cell
response by suppressing inflammatory activity. PD-1 is a 50–55-kDa death protein-1 (PD-1), PD-L1 ligand, and biomolecules able to inhibit
type I transmembrane glycoprotein (Wu et al., 2019a). The ligand or moderate the immune checkpoint proteins, with the emphasis on the
interacting with PD-1, called PD-L1, known also as CD274 or B7-H1, is a invaluable aid of a variety of different biosensing platforms, including
transmembrane protein which is expressed in tumor cells, macrophages, microfluidic devices, biosensors, fiber gratings coated with gold
mast, or dendritic cells. Both PD-1 and PD-L1 belong to the immune nano-shells, as well as plasmonic nanoprobes based on gated resonance
checkpoint protein family. The combination of PD-L1 and PD-1 can energy transfer (gRET) for the cancer diagnosis and treatment. The
inhibit the activation, proliferation and anti-tumor function of CD8+ T sensing systems enabling PD-1/PD-L1 inhibitors’ screening are reviewed
cells, thus enabling the tumor immune escape (Ai et al., 2020). as well. The efficacy of PD-1/PD-L1 inhibitors, being the key acting

Fig. 1. (A) Estimated global burden of cancer in 2018


and 2040 according to World Health Organization
(WHO) (2020) (Licence: CC BY-NC-SA 3.0 IGO); (B)
Classes of immunotherapies. PD-L1 mAb, pro­
grammed cell death ligand 1 monoclonal antibody;
PD-1 mAb, programmed cell death 1 monoclonal
antibody; CTLA4 mAb, monoclonal antibodies tar­
geting cytotoxic T-lymphocyte antigen- 4; IL-2,
interleukin-2; IFN-α, interferon -α; IL-15, inter­
leukin-15; IL-21, interleukin-21; T-Vec, talimogene
laherparepvec; TCR, T cell receptor; CAR T, chimeric
antigen receptor T cells.

2
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

elements in the cancer immunotherapy, has been evaluated based on advanced solid tumors (NCT02964013), Niu et al., (2022) have found
recent investigations utilizing surface plasmon resonance (SPR) and that the humanized immunoglobulin G1 monoclonal antibody, that
homogeneous time-resolved fluorescence (HTRF) techniques. We also binds to TIGIT vibostolimab (MK-7684), with an anti-PD-1 antibody
assess the utility of new theranostic drug nanocarriers, such as the poly pembrolizumab, was well tolerated, and demonstrated antitumor ac­
(lactic-co-glycolic acid) (PLGA) nanoparticles, micelles with functional tivity in patients with non-small-cell lung cancer (NSCLC). Thibaudin
penetrating peptides conjugated with cholesterol and histidine, and et al., (2022) have suggested that the combination of atezolizumab
apoferritin nanocages, as the carriers of small interfering RNAs (siRNAs) (anti-PD-L1) and tiragolumab (anti-TIGIT) in ex vivo evaluation could
for inhibition of PD-L1/PD-1 expression. The potential and challenges of restore function of CD4 and CD8 tumor-infiltrating lymphocytes (TILs)
utilizing PD-L exosomes as the novel therapeutic target are also high­ in microsatellite stable colorectal cancers (MSS-CRC). Hung et al.,
lighted. Furthermore, we discuss the future outlook and potential ap­ (2018) have examined the efficacy and mechanism of dual anti-PD-1
plications of biosensors and exosomes for testing the immune and anti-TIGIT checkpoint blockade in a murine glioma model.
checkpoint blockade, drug development, and cancer treatment. Finally, Another immune checkpoint utilized as the target in combination cancer
we evaluate the prospects of innovative targets for cancer diagnostics immunotherapy is the type I transmembrane glycoprotein, member of
and immunotherapy. the immunoglobulin (Ig) superfamily, that interacts with nectin and
nectin-like proteins, CD96 (TACTILE) (Feng et al., 2023; Liu et al.,
2. PD-1/PD-L1 immune checkpoint blockade 2020). Mittal et al., (2019), tested the anti–PD-1/CD96/TIGIT in a
triple-combination therapy. They have demonstrated that this triple
The development of cancer treatments able to induce an efficient combination is superior in reducing the tumor growth and improving
immune checkpoint blockade by binding PD-1 or PD-L1, is urgently the survival of mice in B16F10 melanoma and CT26 colon carcinoma,
needed to successfully treat cancer. To interrupt the negative control of compared with monotherapy or dual-combination therapy. The combi­
immune response, the brakes of immune checkpoints (i.e., the bridges nation of antibodies against CD96 and PD-1 has also enhanced the delay
PD-1/PD-L1) must be released. While any compound with strong affinity of tumor growth and prolongation of the survival of tumor-bearing mice
to PD-1, able to suppress immune-controlling activity of PD-1, can in (TC-1 tumor-bearing C57BL/6 mouse) (Wang et al., 2022). In a synge­
principle be considered as the potential candidate for a checkpoint in­ neic model of prostate cancer that is resistant to immune checkpoint
hibitor, two classes of monoclonal antibodies (MoAbs), and have been inhibitors (ICIs), inhibition of the binding of vascular endothelial growth
designed to specifically serve for this purpose: the anti-PD-1 and the factor (VEGF) to neuropilin-2 (NRP2) using a mouse-specific anti-NRP2
anti-PD-L1 MoAbs. The antibodies against PD-1 are fully human and monoclonal antibody (mAb) resulted in necrosis and tumor regression
humanized IgG4 and the antibodies against PD-L1 are IgG1 isotypes compared with both an anti–PD-L1 mAb and control immunoglobulin G
with genetically modified Fc fragments (Homet Moreno and Ribas, (Wang et al., 2023). However, de Filette et al. (de Filette et al., 2019) in a
2015; Passiglia et al., 2016). systematic analysis have listed adverse events provoked by single agent
To better understand the scope and challenges of controlling the checkpoint blockade, further reinforced by combined treatment. Patel
immune checkpoints, for the sake of designing biosensing devices et al., (2022) have investigated the role of regulatory B (Breg) cells in the
capable of monitoring the activity of key biomolecules and designer development of severe immune-related adverse events in lung cancer
drugs involved in checkpoint operation, we need to consider first the patients treated with anti-PD-1/PD-L1.
biomedical principles of this autonomous checkpoint functioning. The The immune checkpoint inhibitors, based on monoclonal antibodies
comprehensive background provided in this Section enables better un­ against PD-1 receptor, such as nivolumab and pembrolizumab, and
derstanding of the modifying power, the proteins PD-1, PD-L1, and the those based on the anti-PD-L1 antibodies, such as atezolizumab, avelu­
inhibitor drugs have on the tumor survivability. mab and durvalumab, were successfully approved by the US Food and
The immune checkpoint inhibitors prevent binding of T cells to Drug Administration (FDA) for different types of cancer and commer­
cancer cells (i.e. binding of PD-1 receptors to PD-L1 ligands), and thus, cialized over the last few years (Homet Moreno and Ribas, 2015;
block the immune checkpoints and enable an uninterrupted release of Novotny et al., 2016; Wu et al., 2019a; Xu et al., 2018). According to the
immune attack on cancer cells resulting in the elimination of cancer Web of Science website (https://www.webofscience.com/wos/woscc/­
(Riley et al., 2019; Wu et al., 2019b). Anti-PD-1 antibody binds to PD-1 citation-report/5dddc8d6-e2f9-4c2b-a7a2-62e9a7416f54-959ad3d6
and anti-PD-L1 binds to PD-L1 preventing the binding of PD-1 to its li­ 2023), the publications record concerning the PD-1/PD-L1 immune
gands and PD-L1 to PD-1, releasing the tumor-specific killing ability of T checkpoint inhibitors began to increase from 2014 after the first
cells. Therefore, the introduction of immune checkpoint inhibitors has approval of PD-1/PD-L1 inhibitor and the publications growth rate is
recently become a promising therapy for cancer treatment (Akinleye and still increasing (Fig. 2A). It also indicates that the number of research
Rasool, 2019; Fritz and Lenardo, 2019; Garrett and Collins, 2011; Li groups developing the biosensors for detection of PD-1/PD-L1 immune
et al., 2016; Lipson et al., 2015; Pardoll, 2012; Wu et al., 2019a; Xu et al., checkpoint proteins is only beginning to increase and more de­
2018). The blockade of checkpoints by antibodies against PD-1 and velopments will be published in near future. In Fig. 2 B and Table 1, we
PD-L1 was investigated for cohorts of a large number of patients with summarized the PD-1/PD-L1 inhibitor drugs approved recently, along
melanoma (Bhandaru and Rotte, 2017; Hu-Lieskovan and Ribas, 2017), with the information on targeted cancer, date of approval, and adverse
non-small-cell lung carcinoma (NSCLC) (Aguiar et al., 2017; Passiglia side events.
et al., 2016), renal cell carcinoma (RCC), bladder cancer (BC), Hodgkin
Lymphoma, triple-negative breast cancer (TNBC), and hepatocellular 3. Biosensing technologies for the detection of PD-1/PD-L1
carcinoma (HCC), as well as for hematologic malignancies (Brahmer immune checkpoint proteins
et al., 2012; Homet Moreno and Ribas, 2015; Macek Jilkova et al., 2019;
Philips and Atkins, 2014; Villasboas and Ansell, 2016; Xu et al., 2018). To provide a simple means for monitoring of immune checkpoint
To improve the clinical outcomes for patients, the combination activity, many research groups have recently been actively working on
therapies utilizing a set of different immune checkpoint inhibitors are the development of new analytical methods and biosensing technologies
also frequently applied. The new emerging immune checkpoint target for fast and sensitive determination of checkpoint proteins (Briones
for immunotherapy consists of a T cell immunoreceptor with Ig and et al., 2020; Chou et al., 2018; Reza et al., 2019; Wuethrich et al., 2019).
immunoreceptor tyrosine-based inhibitory motif (ITIM) domains As will be discussed in this section, for the detection of PD-1 and PD-L1
(TIGIT), mainly expressed on natural killer (NK) cells and regulatory T proteins, the microfluidic biosensing platforms, immunosensors, apta­
cells (Tregs) (Zhao et al., 2023; Harjunpää and Guillerey, 2019; Dougall sensors, as well as plasmonic assays, have recently been designed. The
et al., 2017). In the first, in-human phase 1 study of patients with Authors have utilized various techniques, including flow cytometry,

3
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Fig. 2. (A) The records for PD-1/PD-L1 immune checkpoint inhibitors topic according to WoS website; (B) Examples of immune checkpoint inhibitors approved
by FDA.

Table 1
Approved PD-1/PD-L1 inhibitors.
Immune Approved drugs Cancer treatment Approval Treatment-related adverse events Ref.
checkpoint date
target

PD-1 nivolumab melanoma 2014 hypothyroidism, hyperthyroidis, thyroiditis, (Bhandaru and Rotte, 2017;
non-small cell lung cancer 2015 hypophysitis, diabetes mellitus, primary adrenal de Filette et al., 2019;
(NSCLC) insufficiency, Ksienski et al., 2019)
metastatic renal cell carcinoma 2015 dermatitis, colitis
Hodgkin lymphoma 2016
recurrent and metastatic 2016
squamous cell carcinoma of
head and neck (SCCHN)
urothelial carcinoma 2017
pembrolizumab metastatic small cell lung 2018 hypothyroidism, hyperthyroidis, thyroiditis, (de Filette et al., 2019;
cancer hypophysitis, diabetes mellitus, primary adrenal Ksienski et al., 2019;
melanoma 2014 insufficiency, pneumonitis, athralgis, dermatitis Passiglia et al., 2016)
non-squamous non-small cell 2015
lung cancer (NSCLC)
non-muscle invasive bladder 2020
cancer (NMIBC)
cemiplimab-rwlc advanced cutaneous squamous 2018 diarrhea, arthralgia, hypothyroidism, muscle (Ahmed et al., 2019)
cell carcinoma (CSCC) weakness, macularpapular rash, nausea, pruritis,
rash, cough
PD-L1 atezolizumab urothelial carcinoma 2016 hypothyroidism, diabetes mellitus (de Filette et al., 2019)
metastatic non-small cell lung 2016
cancer (NSCLC)
durvalumb locally advanced or metastatic 2017 hypothyroidism (de Filette et al., 2019)
urothelial carcinoma
avelumab Merkel-cell carcinoma 2017 hypothyroidism, hyperthyroidis, diabetes mellitus, (de Filette et al., 2019)
primary adrenal insufficiency
Combination pembrolizumab plus advanced endometrial 2019 hypertension, diarrhea, fatigue, decreased appetite, (Arora et al., 2020; Makker
thearpy lenvatinib carcinoma hypothyroidism, nausea, stomatitis, pain and et al., 2020)
arthralgia, dysphonia
Combination ipilimumab plus irresectable metastatic 2015 hypothyroidism, hyperthyroidis, thyroiditis, (Bhandaru and Rotte, 2017;
therapy nivolumab melanoma hypophysitis, primary adrenal insufficiency, colitis, de Filette et al., 2019)
liver abnormalities, myocarditis

surface enhanced resonance spectroscopy, fluorescence, and electro­ reagents, as well as multiple working channels enabling simultaneous
chemistry for analytical signal measurements (Table 2). measurements for multiple biomarkers detection.
The advantages and disadvantages of different biosensing technol­ Reza et al., (2019) have developed an integrated surface-enhanced
ogies are discussed in detail in subsections 3.1-3.3 and they are also Raman scattering (SERS)- microfluidic platform, with an
summarized in Table 2. Various detection methods used in these tech­ alternating-current electrohydrodynamic (ac-EHD, f = 1.1 kHz and Vpp
nologies and the achieved detection limits are also evaluated in this = 120 mV) mass transport, for the detection of immune checkpoint
Table. proteins (Fig. 3). The gold electrodes were functionalized with graphene
oxide (GO) and hemagglutinin (HA) antibodies using 1-ethyl-3-(3-dime­
thylaminopropyl) carbodiimide/N-hydroxysuccinimide (EDC/NHS)
3.1. Integrated microfluidic systems for immune checkpoint biomarkers chemistry (EDC: 0.2 M; NHS: 0.05 M; mixing volume ratio of 1: 1). Next,
detection the nanoyeast single chain variable fragments (NYscFv), specific to
immune checkpoints PD-1, PD-L1, and LAG-3, tagged with human
The microfluidic devices have demonstrated good sensitivities for influenza hemagglutinin antigen (10 μg/mL), were attached. Owing to
analyzed antigens and significant advantages, such as the system mini­ laborious, long and costly production times of antibody affinity
aturization, simplicity, automation, minimal volume of samples and

4
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Table 2
Biosensing technologies for detecting PD-1/PD-L1 immune checkpoint proteins.
Biosensor Immune Detection method Limit of Real Advantage(s) Disadvantage(s)/challenge(s)/ Ref.
check- detection sample limits of detection
point

Immuno-sensor sPD-L1 surface plasmon resonance 1.75 ng serum sensitive and low concentration in serum and (Hu et al., 2021)
(SPR); /mL samples selective; various body fluids;
cyclic voltammetry (CV); alternative for anti- natural SIBP obtained without
electrochemical impedance bodies; tedious selecting process;
spectroscopy (EIS) strong binding ability tested in diluted serum from
healthy donors and cancer patients
Immuno-sensor PD-L1 surface plasmon resonance 3.29 ng/ human high surface-to- dual recognition; (Huang et al.,
(SPR); mL plasma volume ratio; tested in blood samples collected 2021)
cyclic voltammetry (CV); strong magneto- from healthy donors and cancer
electrochemical impedance optical effect; patients
spectroscopy (EIS); high sensitivity;
atomic force microscope low detection limit;
(AFM) specificity;
template-free method;
excellent electrical
conductivity
Aptasensor PD-L1 flow cytometry 10 cells/ n.a. highly affinite and not tested with real clinical (Yazdian-Robati
mL selective; samples; et al., 2017)
alternative for small population of cancer cells;
antibodies internalization at 37 ◦ C is better
than at 4 ◦ C;
require multiple washing steps
Nanoplasmo-nic PD-L1 gated fluorescence 1.2 nM n.a. very sensitive; not tested with real clinical (Grel et al., 2020)
assay resonance energy transfer simple fabrication; samples;
(gRET) rapid detection could only detect one biomarker;
require modification by antibodies
to increase the specificity of the
system
ExTFG-LSPR sPD-L1 surface plasmon resonance 1 pg/mL PBS probing processes decreased in senitivity when (Luo et al., 2019)
immunosen-sors (SPR) (0.04pM) within the nanometer applied in FBS sample;
5 pg/mL fetal scale; not tested with real clinical
bovine no effect of tempe- samples; detecting of small
serum rature on the sensor biomolecules (sPD-L1, 26 kDa); the
(FBS) response; detection range could be improved
stability of ExTFG; by using graphene oxide (GO) to
extremely specific encapsulate the gold nanoshells
rapid and label-free
detection;
160 nm gold nano-
shells have larger
specific surface area
than 80 nm
Chip-based array PD-1 SERS-microfluidic platform 100 fg/ spiked enable simultaneous complex fabrication process; (Reza et al.,
mL human detection; background removal from the raw 2019)
PD-L1 50 fg/mL serum rapid detection; spectra;
LAG-3 100 fg/ samples multiplexed facilitate simultaneous monitoring
mL detection; and suggest effective therapy
highly sensitive and
specific
alternative for anti-
bodies;
good reproducibility;
excellent storage
stability
sandwich-type sPD-1 SERS platform 6.17 pg/ spiked enable specific straightforward and (Li et al., 2018)
SERS mL human multiplexed environmentally friendly synthesis
platform sPD-L1 0.68 pg/ serum detection; workflow does not require
mL cheap and specific surfactants, high temperature or an
sEGFR 69.86 pg/ detection; organic phase;
mL alternative for anti- developed nanoyeast-scFvs in
bodies fragments of yeast cell walls;
not tested with real clinical
samples
microfluidic PD-L1 flow-proteometric (FAP) 30.5 pM HeLa cells rapid detection; combination a microfluidic-based (Chou et al.,
immunoassay microfluidic platform 2.33 pM MDA-MB- sensitive; system with fluorescence 2018)
231 cells standard-free tech- correlation spectroscopy (FCS) as
patient nique the platform FAP to automatically
breast determine;
tumor for the protein concentrations
tissue <0.13 pM extending the detection
samples time and increasing the flow speed
is necessary;
(continued on next page)

5
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Table 2 (continued )
Biosensor Immune Detection method Limit of Real Advantage(s) Disadvantage(s)/challenge(s)/ Ref.
check- detection sample limits of detection
point

non-specific binding of antibody to


cell or tissue sample may limited
the sensitivity
microfluidic PD-1 alternating-current 5 pg/mL n.a. alternative for anti- not tested with real clinical (Wuethrich et al.,
hemagglutinin PD-L1 electrohydrodynamic (ac- 5 pg/mL bodies; samples; 2019)
(HA) LAG-3 EHD) nanofluidic read-out 50 pg/mL enable multiplexed ac-EHD system generates a
immunosen-sor detection from small nanoscopic fluid flow;
sample volumes; slow mass transport of the analytes,
simultaneous analysis causes slow response time and
of 28 samples; nonspecific interference from the
colorimetric read-out nontarget molecules

Fig. 3. (A) Schematic illustration of the SERS GO ac-EHD immunoassay for immmuno checkpoint blockade biomarkers. (B) Typical SERS spectra and dynamic range
for PD-1. The serum samples spiked with targets were run through the device under an ac-EHD field (f = 1.1 kHz and Vpp = 120 mV). Control experiments were
performed using blank serum samples (0 fg/mL). Adopted from (Reza et al., 2019).

reagents, the Authors have utilized recombinant affinity reagents PD-L1, and 7-mercapto-4-methylcoumarin (MMC) for LAG-3. The
NYscFv, that can be generated quickly, at a low cost, with high speci­ channel without the NYscFv was used as a negative control. The Authors
ficity toward their target antigens and high stability (Grewal et al., have shown that this SERS platform can be used for the detection of
2015). NYscFv designed as the new diagnostic protein capture agent and immune checkpoint markers spiked in human serum down to LOD =
alternative to full length monoclonal antibodies were generated by 100 fg/mL. Thus, due to the sensitive multiplex detection of targets in
mechanical fragmentation, followed by filtration to produce yeast-cell human serum samples, this method offers a high potential for fast cancer
wall fragments of varying sizes, with the single-chain variable frag­ screening. Also, the successful application of NYscFv in a biosensing
ments (scFvs) complex remaining intact (Grewal et al. 2015, 2016). platform provides a promising low-cost alternative in comparison to the
Then, the samples (20 μL) containing immune checkpoint antigens expensive monoclonal antibodies used in current methods such as the
(PD-1 (100 fg/mL to 1 ng/mL), PD-L1 (50 fg/mL to 100 pg/mL), and ELISA.
LAG-3 (100 fg/mL to 1 ng/mL) were injected into different channels of Biosensing technologies based on integrated microfluidic platforms
the ac-EHD device. Subsequently, the captured antigens were detected have also been developed for the determination of PD-L1 protein.
using three different SERS nanotags consisting of gold nanoparticles and Recently, Chou et al., (2018) designed a flow-proteometric microfluidic
Raman reporters, including mercaptobenzoic acid (MBA, 5 μL of 1 mM) platform for analyzing proteins (FAP). The concentration of PD-L1 was
for PD-1, 5,5′-dithio-bis-[2-nitrobenzoic acid] (DTNB, 10 μL of 1 mM) for determined in vitro in cultured cancer cells (HeLa and MDA-MB-231)

6
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

and in vivo in breast tumor tissue samples from patients. The cells were conjugated to the anti-hemagglutinin antibody functionalized electrode
labeled first with Alexa Fluor 647-conjugated anti-PD-L1 antibody and surface. The sample was added together with horseradish peroxidase
then the cell lysate was collected and injected into the FAP platform for (HRP)-conjugated secondary antibody against the immune checkpoint
PD-L1 detection. The Authors have also performed a negative control proteins in PBS. The analytical color change signal was obtained after
test with isotype IgG-Alexa647 antibody to demonstrate a nonspecific the enzymatic oxidation of TMB in the presence of HRP. The TMB color
binding with PD-L1 protein. The individual photon burst signal from a change from transparent to yellow (maximum absorbance wavelength
fluorescence dye was identified and counted as the analytical signal. The λmax = 450 nm) was observed after reaction quenching with sulfuric
concentrations of PD-L1 determined in channels of the microfluidic acid. The limits of detection (LODs) for PD-1, PD-L1, and LAG-3 immune
biosensor, were 30.5 pM and 2.33 pM, for HeLa and MDA-MB-231 cells checkpoint proteins, of 5 pg/mL, 5 pg/mL, and 50 pg/mL, were ach­
lysates, respectively. From these values, the original PD-L1 concentra­ ieved, respectively. The proposed biosensor offers an advantage of the
tions in single cells can be determined, on the bases of the numbers of parallel detection of several immune checkpoint markers and therefore,
cells being analyzed (Ncells = 5×104), and single cell volumes of 4.7 pL is capable of detecting a multitude of biomarkers at once.
and 5.7 pL, for HeLa and MDA-MB-231 cells, respectively. Considering
the necessary dilutions of the cell lysates to prevent microchannel 3.2. Optical systems for immune checkpoint biomarkers detection
clogging (9x and 5x dilutions, respectively), the cellular PD-L1 con­
centrations of 357 and 12.3 nM, for HeLa and MDA-MB-231 cells, were A new optical biosensing system based on a sandwich-type SERS
obtained. Here, the cellular concentration of PD-L1 is defined as the total platform was developed by Li et al., (2018). The Authors have utilized
number of moles of PD-L1 recovered during cell lysis from the cell the nanoyeast-scFvs (NYscFv) conjugated with streptavidin-coated
membrane and cytosol, divided by the single cell volume. The results of magnetic beads (MBs) for multiplexed detection of soluble cancer pro­
measurements performed in tissues were consistent with those obtained tein biomarkers including soluble programmed death 1 (sPD-1), soluble
by conventional immunohistochemistry (IHC) staining. The concentra­ programmed death-ligand 1 (sPD-L1) and soluble epidermal growth
tions of PD-L1 in original tissues of patients under study yielded values factor receptor (sEGFR). The SERS tags consisted of the anisotropic gold
in the range from 0.73 to 5.35 nM for patient A and patient B, respec­ (Au)-silver (Ag) (Au-Ag) alloy nanoboxes (NBs) used for the signal
tively (Fig. 4). Thus, the proposed flow-proteometric microfluidic plat­ enhancement, Raman reporters: 4-mercaptobenzoic acid (MBA), 5,
form offers a high sensitivity and a wide dynamic range. Other 5-dithiobis(2-nitrobenzoic acid) (DTNB), 2,3,5,6-tetrafluoro-4-mercap­
advantages of the method include measurement simplicity, speed, and tobenzoic acid (TFMBA), which generate unique Raman signals at
reliability of protein determination in cells and tissues. 1585 cm-1, 1335 cm-1 and 1376 cm-1, respectively, and anti-PD-1,
The detection of immune checkpoints, including PD-1, PD-L1, and anti-PD-L1 and anti-EGFR polyclonal antibodies, used as the affinity
LAG-3, was also reported by Wuethrich et al., (2019). They have reagents (Fig. 5). The biosensor developed has enabled detection of
developed a multiplexed immune checkpoint biosensor (MICB), sPD-1, sPD-L1, and sEGFR, with the limits of detection (LOD) of 6.17
designed on the principle of a sandwich immunoassay with in situ pg/mL, 0.68 pg/mL, and 69.86 pg/mL, respectively. The tests in spiked
alternating-current electrohydrodynamic (ac-EHD) nanofluidic mixing human serum samples have also been performed with recovery rates
and sensitive colorimetric read-out, based on the enzymatic conversion between 82.99% and 101.67%. Due to the extremely sensitive detection
of 3,3′,5,5′-tetramethylbenzidine (TMB). The new class of low-cost and of cancer biomarkers, this new SERS platform may be able to help in
stable protein capture agents, nanoyeast single chain variable fragments early and timely diagnosing and cancer therapy.
(scFvs) specific to immune checkpoints PD-1, PD-L1, and LAG-3 were Very sensitive surface plasmon resonance (SPR) transduction based

Fig. 4. (A) Automated single-molecule-detection platform. (B) Immunohistochemistry (IHC) staining results and (C) PD-L1 concentration measurement by FAP
platform for PD-L1 expression in breast cancer tissue from patient A and patient B. Adopted from (Chou et al., 2018).

7
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Fig. 5. Schematic illustration for the detection of sPD-1, sPD-L1 and sEGFR using SERS platform. Adopted from (Li et al., 2018).

biosensor for the detection of soluble PD-L1 (sPD-L1) immune check­ down to 1.2 nM (Grel et al., 2020) (Fig. 6). The adsorption of PD-L1
point biomarker has been designed by Luo et al., (2019). The on AuNP was observed due to the high affinity of PD-L1 to the surface
anti-sPD-L1 monoclonal antibodies (anti-sPD-L1 MAbs) were linked to coating on the AuNP. It has been found that the high affinity of PD-L1 to
the surface of the sensor by the staphylococcal protein A (SPA). The SPR AuNP@Cit is associated with: (i) supramolecular interactions of amino
immunosensor built on an excessively tilted fiber grating (ExTFG), acids of the protein with citrates in AuNP coating, due to hydrogen
coated with large-sized gold nano-shells (~160 nm) capped with bonding, (ii) partial replacement of citrates from AuNP surface and
cysteamine, was able to detect sPD-L1 down to an extremely low direct bonding of PD-L1 with Au surface atoms via Au-S covalent
detection limit, LOD = 1 pg/mL (0.04 pM) and LOD = 5 pg/mL, in bonding, due to some of the six cysteine residues present in the structure
phosphate-buffered solution (PBS) and fetal bovine serum (FBS) sam­ of PD-L1; (iii) cation-mediated bridge formation, and (iv) strong elec­
ples, respectively. Although, the immunosensor was highly specific to trostatic screening of negative charges of the components by relatively
sPD-L1 molecules, the detection range was not satisfactory. The sensi­ high ionic strength of the 50 mM buffer solution used.
tivity of the biosensor could be further improved by using graphene The gRET method has the advantages of simple detection mechanism
oxide (GO) to encapsulate the gold nano-shells before immobilization on and convenient operation while obtaining a very sensitive detection
the fiber grating. The reduction of the core/cladding diameter of ExTFGs limit in the buffer solution in absence of interferent proteins. However,
could also be applied for improving the sensitivity of the proposed in biological matrix samples such as a serum, the implementation of
ExTFG-LSPR sensors. This genosensor’s advantages are also associated antibodies against PD-L1 attached to the gold nanoparticles have to be
with the determination speed and label-free detection of sPD-L1 anti­ applied for improvement of the method specificity toward the detection
gens, thus offering a potential application in cancer detection. of biomarkers in the body fluids. Also, for the simultaneous detection of
Aptamers against transmembrane protein PD-L1 have also been immune checkpoint proteins, different specific antibodies should be
synthesized to enable controlling the immune checkpoints. Yazdian-­ immobilized onto gold nanoparticles surface.
Robati et al., (2017) have developed new DNA aptamers (Apt5 and
Apt33) against the transmembrane protein PD-L1 using Protein-SELEX 4. Inhibitors for PD-1/PD-L1 immune checkpoint blockade
method based on the Ni2+ complex with nitrilotriacetic acid (Ni-NTA)
adsorbed on agarose magnetic beads. The aptamer with better binding The binding properties of monoclonal antibodies (mAbs) against PD-
affinity to the target (Apt5) was further investigated using flow cytom­ 1/PD-L1, which are the key factors in the immune checkpoint inhibition,
etry. The synthesized aptamer, labeled with ATTO 647 N red-emitting can conveniently be investigated using the surface plasmon resonance
fluorescence dye, was applied for the detection of A2780 human (SPR) technique. Such investigations have been performed by several
ovarian carcinoma cell line. This fluorescent aptasensor enabled tumor groups (Brown et al., 2020; Ghiotto et al., 2010; Luan et al., 2016; Tan
cells detection with LOD 10 cells/mL and linearity in the range from 50 et al., 2018)
cells/mL to 1000 cells/mL, which is comparable to the results of other
analytical methods for PD-L1 positive cancer cells determination, such 4.1. Antibodies as immune checkpoint inhibitor
as the dual-aptamer target binding strategy, localized surface plasmon
resonance (LSPR), fluorescence, and electrochemical methods. There­ Recently, Brown et al. (Brown et al., (2020) have investigated the
fore, the proposed method has a high potential for use in early stages of effect of chip type (flat or 3D-hydrogel) on the SPR-derived binding rate
cancer development. Furthermore, the aptamers are promising alter­ constants and affinities of monoclonal antibodies (mAbs) against pro­
natives to antibodies due to the easy synthesis and such features as the grammed cell death protein 1 (PD-1) to their specific target antigens.
unique tertiary structure with high specificity and affinity towards The capture surface of chip was prepared by amine-coupling of anti­
target molecules, no immunogenicity, no toxicity, resistance to the high bodies onto planar surfaces such as carboxymethyldextran exhibiting
temperature, long storage time, as well as the ease of modification. the thickness < 5 nm (CMD-P), as well as a nearly flat surface such as
polycarboxylate hydrogel exhibiting the coating thickness about 30 nm
3.3. Plasmonic systems for immune checkpoint biomarkers detection (HC-30M) or 3D-hydrogel such as carboxymethyldextran hydrogel,
exhibiting the coating thickness about 200 nm (CMD-200M). The Au­
Recently, for the highly sensitive determination of proteins, a bio­ thors have utilized in their investigations a lower throughput Biacore 8K
sensing technology, based on the modulation of resonance energy SPR instrument and a high-throughput Carterra LSA platform. A variety
transfer (RET), was proposed (Grel et al., 2020; Stobiecka, 2014; Sto­ of epitope coverage and ability of mAbs to block the direct interaction
biecka and Chalupa, 2015). The method was based on the modulation of between PD-1 and PD-L1 proteins was also studied. The Authors have
RET from a donor fluorescent dye molecule to a plasmonic gold nano­ screened over thirty mAbs including camrelizumab (camre), cemiplimab
particle (AuNP), acting as the acceptor, through a sub-monolayer (cemip), pembrolizumab (pembro), nivolumab (nivo), and sintilimab
adsorption film formed by PD-L1 protein on a AuNPs. Voids in the (sinti). They have shown, that only three of the antibodies (mAb05,
protein film acted as the gates for RET and enabled detection of PD-L1 mAb12, and mAb30) were unable to block the binding to PD-L1 (Fig. 7).

8
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Fig. 6. (A) Principle of gated resonance energy


transfer (gRET) (drawn not to scale); (B) Dependence
of the gRET efficiency for energy transfer from FITC
to AuNP@Cit on PD-L1 concentration showing satu­
ration at higher PD-L1 concentrations; (C) Details of
the limit of detection (LOD) determination using 3σ
method (LOD = 1.2 nM); CPD-L1 [nM]: (1) 0, (2) 1.27,
(3) 3.17, (4) 6.3, (5) 12.7, (6) 25.3, (7) 38, (8) 51, (9)
63.0; CFITC = 66.7 nM; CAuNP@Cit = 2.02 nM; λex =
495 nm. Adopted from (Grel et al., 2020), Creative
Commons Attribution (CC BY) license (http://cre
ativecommons.org/licenses/by/4.0/).

Fig. 7. Network plots depicting the epitope clusters deduced from binning a panel of 31 anti-PD-1 mAbs. (A) The coloured bins represented by a family of mAbs
sharing an identical blocking profile, (B) PD-L1 blockade by mAbs (green = blocks, red = does not block). Adopted from (Brown et al., 2020), Creative Commons
Attribution License.

These label-free investigations enable to gain deeper insights into the important core for developing the PD-1 inhibitors. These investigations
mechanism of action of new drugs under development for cancer indicate that instead of monoclonal antibodies, other organic molecules
treatment. may be used and applied as the programmed cell death-1inhibitors in
cancer therapy. It opens a new way for the design and synthesis of a new
4.2. Small molecules and peptides as immune checkpoint inhibitors class of drugs.
Han et al., (2018) have used a SPR technique for the investigation of
Due to the increasing number of cancer cases, the exploration of new the affinity and competitive inhibition of nine caffeoylquinic acid
methods for tumor treatment is of great interest. Recently, in addition to compounds (CQAs) against PD-1/PD-L1. They have found four small
the mAbs utilized as the PD-1 and PD-L1 inhibitors, researchers have molecules, including 1-CQA, 3-CQA, 4-CQA, and 5-CQA, that could act
also developed new peptides, organic molecules, and natural products as the PD-1/PD-L1 inhibitors. The 3-CQA, 4-CQA and 5-CQA inhibitors
against the immune checkpoints. Such studies have been performed by revealed micromolar inhibition with low IC50 values equal 36.6, 38.3,
Liu et al., (2016). The Authors have discovered new small molecule and 45.6 μM, respectively, and 1-CQA has shown inhibition of
inhibitors of PD-1 by applying a homogeneous time-resolved fluores­ PD-1/PD-L1 with somewhat higher IC50 value of 87.3 μM (Fig. 8). Thus,
cence (HTRF) technique for investigation of the inhibitive activity of the SPR technology, due to its outstanding properties, such as the speed,
sulfamethizole and sulfamethoxypyridazine synthetic compounds. They sensitivity, and label-free measurements, has been explored for effective
have employed the N,N-dimethylcarbamate and other alkyl-substituted and rapid screening of small molecules to find the immune checkpoint
amines and resorcinol (a dihydroxyl phenol at meta-position) as the inhibitors and, thereby, SPR has become the method utilized in cancer

9
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Fig. 8. (A-B) The example of competitive inhibition of two CQAs (1-CDQA (A) and 4-CDQA (B) on PD-1/PD-L1. CPD-1 = 392 nM; (C) Structures of 1-CQA, 3-CQA, 4-
CQA, 5-CQA; (D) The IC50 curves of small molecules including 1-CQA, 3-CQA, 4-CQA, 5-CQA on PD-1/PD-L1. Ferulic acid is a negative control. Adopted from (Han
et al., 2018).

treatment. the host and grow freely despite the presence of T cells (Grel et al., 2020;
A phage display biopanning strategy for screening of small peptide- Santarpia and Karachaliou, 2015). Therefore, developing brakes capable
based anti-PD-L1 inhibitors (CLP001, CLP002, CLP003, CLP004) has of targeting the PD-1/PD-L1 pathway is highly desired and has become
been proposed by Cheng’s group (Liu et al., 2019). This method relies on the most extensively researched strategy for cancer treatment. Also, the
the expression of peptide or protein sequence by each phage in phage new technologies for immune checkpoint brakes delivery are urgently
library. The strategy developed is based on binding of the peptide under needed. In addition to the anti-PD-L1 antibodies used as the brakes for
study to specific residues on a target protein (PD-L1), and subsequently the direct blocking of PD-L1 protein, an alternative therapy based on
blocking the protein’s interaction with its PD-1 receptor. The binding nanodelivery of small interfering RNAs (siRNAs) to cancer cells has
affinities of peptides for human PD-L1 protein were determined using recently been applied. For a more effective targeted delivery system and
surface plasmon resonance technique. The system was based on the less toxic cancer treatments, the functionalized nanoparticles have been
PD-L1 protein covalently immobilized by amine coupling method with used (Kosmides et al., 2017; Kwak et al. 2017, 2019; Li et al., 2019b). In
dextran hydrogel functionalized with COOH groups onto a gold chip. Table 3 the most important features of nanocarriers and guiding prin­
The equilibrium dissociation constants (KD), determined for CLP001, ciples for their improvement/creation are listed.
CLP002, CLP003 and CLP004 peptides bound to a human PD-L1 protein A dual immunoswitch, based on an 80 nm iron-dextran particle
were: 534, 366, 117, and 544 nM, respectively. The antitumor activity of platform coated with anti-PD-L1 and anti-4-1BB antibodies, has been
peptides was evaluated using the CT26 colorectal tumor-bearing mice developed by the Schneck’s group (Kosmides et al., 2017). These
and the tumor penetration capability in MDA-MB-231 cells by nanoparticles were able to switch off the immunosuppressive PD-L1
anti-PD-L1 peptides was also investigated. The phage display procedure pathway on tumor cells and switch on the 4-1BB pathway on CD8+ T
was successfully applied for the discovery of small peptide-based cells causing inhibition of tumor growth and are more effective than
checkpoint inhibitors; thus, it has a potential for application in cancer intratumoral injection of soluble antibodies. These immunoswitch
therapy. The developed CLP002 peptide has been found to bind specif­ nanoparticles enhance the efficacy of two immunotherapies and are thus
ically to PD-L1 protein and due to the PD-1/PD-L1 interaction blocking, more effective at low doses in cancer therapy.
has exhibited a better tumor penetration in a 3D tumor spheroid model The simultaneous silencing of PD-1 and PD- L1 expressions on
in comparison with the anti-PD-L1 antibody. Furthermore, it inhibited cytotoxic T cells (CTLs) and colon tissues, respectively, have been pro­
the tumor growth and increased the survival of CT26 mice model. posed by Kwak et al., (2019). The Authors have used the poly
(lactic-co-glycolic acid) (PLGA) nanoparticles loaded with siRNAs
5. Nanoparticle carriers for drug delivery technologies to against PD-1 and its ligand PD-L1 gene (PD-1 siRNA/PD-L1 siR­
suppress PD-1/PD-L1 activity NA@PLGA NPs) for codelivery of PD-1 and PD-L1 siRNAs. The Authors
have found that the co-inhibition of PD-1 and PD-L1 results in enhanced
The expression of PD-L1 protein on the surface of a variety of cancer antitumor effects in comparison with a single silencing of PD-1 or PD-L1
cells causes the inhibition of kinase signaling pathways upon binding of alone, indicating that the concerted inhibition is preferable (Fig. 9). The
these ligands to the immune cell PD-1 receptors and results in sup­ apoptosis process was monitored by the TUNEL staining assay.
pressing of the T cells activity and taming of immune response. In this A nanocarrier system for co-delivery of PD-L1 siRNA (siPD-L1) and
way, cancer cells expressing PD-L1 can easily evade immune reaction of an 1-methyl-DL-tryptophan (1 MT) indoleamine 2,3-dioxygenase (IDO)

10
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Table 3
Advantages and guiding principles of nanocarriers to suppress PD-1/PD-L1 activity.
Nanoparticles Advantages Guiding principles for improvement/creation of Ref.
nanoparticles/disadvantages

stem cell membrane (SCM) camouflaged - efficient loading of DOX and PD-L1 siRNA - good hydrophilicity, biocompa-tibility, stability and (Mu et al.,
polydopamine (PDA) nanoparticles carrying via π–π stacking interactions, biodegrade-bility of dopamine and spontane-ously 2021)
doxorubicin (DOX) and PD-L1 siRNA - - reduction of the DOX toxicity, polymerization to polydo-pamine (PDA) nanoparticles
PDA–DOX/siPD-L1@SCM NPs - - synergistic therapeutic effect, (NPs),
- good compatibility with blood, - large number of catechol and amino functional groups in
PDA for binding many functional molecules to the
surface,
- loss of DOX during the physical extrusion process,
80 nm iron-dextran nanoparticles with the - simultaneously block the inhibitory - combinatorial immunotherapy for cancer treatment as (Kosmides
antibodies against 4-1BB (a co-stimulatory checkpoint PD-L1 signal and stimulate T cells promising success in clinical trials, et al., 2017)
receptor found on the effector T cells) and via the 4-1BB co-stimulatory pathway, - use of nonspecific immunomodulators requires high
antibodies against PD-L1 (found on the cancer - significantly delay tumor growth, doses of the drugs and results in significant off-target side
cells) - extend survival in multiple in vivo models of effects,
murine melanoma and colon cancer,
- increased density, specificity, and in vivo
functionality of tumor-infiltrating CD8+ T
cells checkpoint blockade efficacy,
- synergy between the two immunotherapies,
- effective at low doses,
- target two different cell types in the tumor
microenvironment,
Poly (lactic-co-glycolic acid) (PLGA) nanoparticles - significant tumor growth suppression and - low target specificities, very long half-lives, risk of an (Kwak et al.,
loaded with PD-1 and PD-L1 siRNA long-term tumor inhibition in colon cancer autoimmune response, and limitations of the antibody 2019)
by co-silencing of PD-1 and PD-L1, manufacturing process,
- co-inhibition of PD-1 and PD-L1 exhibited - less likely to respond to single checkpoint blockade,
the enhanced antitumor effects,
siPD-L1 with a polymeric carrier (dubbed “pd”) - suppression of the expression of PD-L1, - specific toxicity of the CLPEI/DS (dubbed “pd”) to a (Kwak et al.,
consisting of disulfide-cross-linked polyethy- - attenuation the expression of tumor-specific group of cancer cells that express CD146, 2017)
leneimine (CLPEI) and dermatan sulfate (DS) - genes in B16F10 cells, - high target specificity of siRNA,
siPD-L1/pd - suppression of melanoma growth in immune-
compromised nude mice,

inhibitor for PD-1/PD-L1 blockade has been developed by Li et al., observed suppression of T cell activation in a draining lymph node and
(2019b). The tumor-targeted system was based on micelles with func­ PD-L1-deficient tumor cells. Chen et al., (2018) have shown that the
tional penetrating peptides (Lin TT1, sequence: AKRGARSTA) conju­ exosomal PD-L1 suppresses anti-tumour immunity systemically and the
gated with cholesterol and histidine 7 (Chol-HHHHHHH-AKRGARSTA, circulating exosomal PD-L1 could become a predictor for the clinical
CHL). The antitumor effect of nanoparticles was evaluated in vitro and in outcomes of anti-PD-1 therapy. Moreover, the exosomal PD-L1 can be
vivo. considered as a negative prognostic factor for pancreatic ductal adeno­
The suppression of PD-L1 expression in melanoma cells has also been carcinoma (PDAC) patients (Lux et al., 2019). Because of the exosomes
achieved by delivering siPD-L1 with a polymeric nanocarriers consisting significant cargo, the tumor-derived exosomes could potentially be used
of disulfide-crosslinked polyethyleneimine (CLPEI) and dermatan sul­ to form a pool of tumor antigens to stimulate the anti-tumor response
fate (DS) (siPD-L1/pd) by Kwak et al., (2017). This nanodrug was (Xie et al., 2019).
applied to the melanoma growth in immune-compromised mice and the Therefore, targeting of the exosomal PD-L1 expression can become a
interferon-γ (IFN-γ)-challenged B16F10 melanoma cells. A successful novel therapeutic approach in future cancer treatment. Li et al., (2019a)
suppression of PD-L1 expression in B16F10 melanoma growth in have demonstrated that the level of exosomal PD-L1 in non-small cell
C57BL/6 immune-competent mice was also reported. lung cancer (NSCLC) patients was higher than in healthy controls and
correlated positively with tumor size, positive lymph node status, distant
6. Exosomal PD-L1 as a novel therapeutic approach in cancer metastasis and advanced TNM stage (Fig. 10). Exosomes were isolated
treatment according to manufacturer protocol from serum collected from blood
samples using the commercial kit (Total Exosome Isolation Kit) by
Exosomes are extracellular vesicles (30-100 nm in diameter) that are centrifugation.
released from cells and contain a rich variety of bioactive molecules
dependent on the type and state of the cell of origin. Recently, several
research groups have discovered that the PD-L1 is also expressed on the 6.1. Detection of exosomal PD-L1 using optical biosensors
surface of exosomes released from different kinds of cancer cells (Chen
et al., 2018; Kim et al., 2019; Poggio et al., 2019; Song et al., 2019; Recent endeavor to elucidate the mechanisms of cancer metastasis
Stobiecka, 2015; Zhang et al., 2023b). Flow cytometry and immuno­ has led to the discovery of exosomal intercellular communication (Sto­
fluorescence results suggested that PD-L1 is present not only on the biecka, 2015; Chung et al., 2020) and a wealth of cancer biomarkers (Li
surface of vesicles but also within the vesicle-like structures (Xie et al., et al., 2020) carried by exosomes secreted by cancer cells. Hence, the
2019; Poggio et al., 2019). The level of PD-L1, carried on the exosome detection of exosomal proteins becomes a new means of non-invasive
surface, was also associated with tumor progression in a variety of cancer screening, staging, and monitoring. The detection of exosomal
cancers including melanoma (Gyukity-Sebestyén et al., 2019), breast proteins, such as the exosomal epidermal growth factor receptor (EGFR)
cancer (Xie et al., 2019), and head and neck squamous cell carcinoma and PD-L1, using the intensity-modulated compact SPR biosensors, was
(HNSCC) (Theodoraki et al., 2018). It was also shown that exosomal reported by Liu and colleagues (Liu et al., 2018). The exosomes were
PD-L1 secreted from cancer cells has immunosuppressive effects (Tang isolated from human serum samples using the exosome kit and the ob­
et al., 2020; Morrissey and Yan, 2020). Poggio et al., (2019) have tained exosome pellets were then resuspended in PBS buffer. The SPR
biosensor was based on a gold biochip modified with a mixture of

11
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Fig. 9. (A) Simultaneous silencing of PD-1 and PD-L1 by systemic delivery of siRNA for suppression of colon tumor growth; (B) In vivo antitumor effects of siR­
NA@PLGA NPs. TUNEL staining assay demonstrating apoptotic cells in the MC38 tumor sections from the injected mouse groups. Scale bar = 50 μm. Adopted from
(Kwak et al., 2019).

methyl-polyethylene glycol-thiol, biotinylated-polyethylene glyco­ signal, by utilization of an efficient DNA amplification based on in situ
l-thiol, NeutrAvidin, and antibodies, including biotinylated anti-EGFR, hyperbranched rolling circle amplification (HRCA). During the HRCA
biotinylated anti-PD-L1, and biotinylated anti-IgG. The exosomal pro­ process, the hydrogen ions are sustainably released along with the
teins were detected by incubating exosomes on the biochip surface. The continuous addition of nucleotides caused the pH of the medium
Authors have demonstrated better detection sensitivity of compact SPR lowering and disassembly of PVP@HRP@ZIF-8 at a mildly acidic envi­
biosensor than ELISA, while providing similar sensing accuracy. They ronment, followed by the release of HRP. A high electrochemical
have shown that ELISA was not able to detect exosomal PD-L1 levels in response was observed in the presence of PD-L1+ exosomes, during
serum samples. The EGFR and PD-L1 biomarkers for cancer diagnostics HRP-catalyzed oxidation of o-phenylenediamine (OPD) with the addi­
were detected in cell culture medium and serum samples from tion of hydrogen peroxide (H2O2). The proposed method was also
non-small-cell lung cancer (NSCLC) patients and normal controls enabled to detect the elevated levels of circulating PD-L1+ exosomes in
(Fig. 11). undiluted serum samples from healthy controls, nonmetastatic breast
A simple analytical method for determination of exosomal PD-L1 cancer patients and metastatic breast cancer patients.
directly from human serum samples was proposed by Pang et al.,
(2020). The system was based on Fe3O4@TiO2 nanoparticles and
6.3. Exosomes as a drug delivery carriers
anti-PD-L1 antibody-modified Au@Ag@MBA nanoparticle SERS tags.
The method allows to detect a single programmed death ligand-1 posi­
Recently it was also shown that the exosomes due to their low
tive (PD-L1+) exosome/μL within 40 min. The method also enabled to
toxicity, high bioactivity, an innate stability, low immunogenicity and
distinguish the early and late nonsmall cell lung cancer (NSCLC) patients
excellent tissue/cell penetration capacity as well as biocompatibility
from healthy controls. Human serum samples were filtrated with a 0.22
become a promising drug delivery carriers (Zhang et al., 2023a; Lee
μm syringe-driven filter, incubated with the nanoparticles for exosome et al., 2022; Sen et al., 2023; Chen et al., 2021; Kar et al., 2023).
isolation, and labeled with SERS tags.
Mu et al., (2021) have developed mesenchymal stem cell membrane
(SCM)-derived vesicles coated polydopamine (PDA) nanoparticles (NPs)
6.2. Detection of exosomal PD-L1 using electrochemical biosensors as an efficient tumor-targeting delivery platform. The nanoparticles
have encapsulated doxorubicin (DOX) chemotherapeutic drug and
Cao et al., (2020) have detected PD-L1+ exosomes with a low limit of siRNA targeting PD-L1 (siPD-L1) loaded onto the surface of PDA NPs via
detection 334 particles/mL using porous nanomaterials consisted of π–π stacking interactions (PDA-DOX/siPD-L1@SCM NPs). The Authors
horseradish peroxidase (HRP) encapsulated into zeolitic imidazolate have shown that the PD-L1 protein on the surface of PC-3 cells was
framework-8 (ZIF-8) coating by polyvinylpyrrolidone (PVP), effectively suppressed by codelivered PD-L1 siRNA and the treatment of
PVP@HRP@ZIF-8. The Authors have attained the enhancement of the mice with PDA–DOX/siPD-L1@SCM effectively inhibit the growth of

12
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Fig. 10. (A) TEM image of exosomes (black arrows)


isolated from NSCLC patients, (B) Schematic diagram
of the formation and secretion of exosomes with PD-
L1 by tumor cells, (C-D) Quantitative analysis of
exosomal PD-L1 (Exo-PD-L1) levels (pg/mL serum)
(C) and relative Exo-PD-L1 levels (pg/mg exosomal
protein) (D), among healthy individuals (n = 27),
stage I–II (n = 57) and III/IV (n = 28) NSCLC pa­
tients; *p<0.05; ***p<0.001; #p<0.05; ##p<0.01.
Adopted from (Li et al., 2019a), Creative Commons
Attribution 4.0 International License (http://creati
vecommons. org/licenses/by/4.0/).

Fig. 11. (A) Setup of compact SPR biosensor (left)


and the photo and schematic diagram of the biochip
(right); (B) Sensing mechanism of compact SPR
biosensor; (C) Representative real-time response
curve of compact SPR biosensor detecting exosomal
PD-L1 in serum sample from a Stage III lung cancer
patient; (D) Expression of exosomal PD-L1 in serum
samples measured by compact SPR biosensor. 50 μL
serum sample was used in all measurements. n = 5, *:
p < 0.05). Adopted from (Liu et al., 2018).

13
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

prostate cancer (PCa) bone metastases in nude mice. the National Science Centre, Poland, Grant No. 2019/34/E/ST4/00281
(to S.J.) is also acknowledged.
7. Conclusions and future outlook of immune checkpoint
blockade in cancer treatment References

The remarkable immune checkpoint inhibitors, developed recently Aguiar Jr., P.N., De Mello, R.A., Hall, P., Tadokoro, H., Lima Lopes, G., 2017. PD-L1
expression as a predictive biomarker in advanced non-small-cell lung cancer:
to control the organism’s own immune responses, have become the key updated survival data. Immunotherapy 9 (6), 499–506.
elements of the emerging methods of cancer immunotherapy. In parallel Ahmed, S.R., Petersen, E., Patel, R., Migden, M.R., 2019. Cemiplimab-rwlc as first and
with these achievements, extensive works have been carried out to only treatment for advanced cutaneous squamous cell carcinoma. Expet Rev. Clin.
Pharmacol. 12 (10), 947–951.
design and test novel biosensors to monitor and screen checkpoint in­ Ai, L., Chen, J., Yan, H., He, Q., Luo, P., Xu, Z., Yang, X., 2020. Research status and
hibitors, and to detect a range of cancer biomarkers to enable quick outlook of PD-1/PD-L1 inhibitors for cancer therapy. Drug Des. Dev. Ther. 14,
evaluation of the cancer treatment. However, there are many factors 3625–3649.
Akinleye, A., Rasool, Z., 2019. Immune checkpoint inhibitors of PD-L1 as cancer
implicated in the immune checkpoint inhibitors response including therapeutics. J. Hematol. Oncol. 12 (1), 92.
circulating immune factors (diverse white blood cells (WBCs)), Arora, S., Balasubramaniam, S., Zhang, W., Zhang, L., Sridhara, R., Spillman, D.,
emerging germline genetic traits, (human leukocyte antigen class I Mathai, J.P., Scott, B., Golding, S.J., Coory, M., Pazdur, R., Beaver, J.A., 2020. FDA
approval summary: pembrolizumab plus lenvatinib for endometrial carcinoma, a
(HLA-I) complex), phenotypic host factors (obesity, cholesterol, gender),
collaborative international review under project orbis. Clin. Cancer Res. 26 (19),
environmental factors (medications), human gut microbiota (Gunjur 5062–5067.
et al., 2022). The blocking of immune checkpoints by monoclonal an­ Asadujjaman, M., Cho, K.H., Jang, D.-J., Kim, J.-E., Jee, J.-P., 2020. Nanotechnology in
tibodies has been particularly effective in improving clinical outcomes the arena of cancer immunotherapy. Arch Pharm. Res. (Seoul) 43, 58–79.
Bhandaru, M., Rotte, A., 2017. Blockade of programmed cell death protein-1 pathway for
for patients with some types of cancers but there is still a long way to the treatment of melanoma. J Dermatol Res Ther 1 (3), 1–11.
attain the expected high efficacy of immunotherapy for most cancers. In Brahmer, J.R., Tykodi, S.S., Chow, L.Q.M., Hwu, W.-J., Topalian, S.L., Hwu, P., Drake, C.
view of the assessment of the progress made in the development of G., Camacho, L.H., Kauh, J., Odunsi, K., Pitot, H.C., Hamid, O., Bhatia, S.,
Martins, R., Eaton, K., Chen, S., Salay, T.M., Alaparthy, S., Grosso, J.F., Korman, A.J.,
PD-1/PD-L1 pathway-based anticancer immunotherapeutic methods, Parker, S.M., Agrawal, S., Goldberg, S.M., Pardoll, D.M., Gupta, A., Wigginton, J.M.,
performed in this work, it is evident that mAbs against PD-1/PD-L1 2012. Safety and activity of anti–PD-L1 antibody in patients with advanced cancer.
demonstrate a great efficacy in cancer treatment in case of cancers, N. Engl. J. Med. 366 (26), 2455–2465.
Briones, J., Espulgar, W., Koyama, S., Takamatsu, H., Tamiya, E., Saito, M., 2020. The
such as melanoma, hepatocellular carcinoma, Hodgkin’s lymphoma, Future of Microfluidics in Immune Checkpoint Blockade. Cancer Gene Therapy.
and NSCLC. We envision that further development of the analytical Brown, M.E., Bedinger, D., Lilov, A., Rathanaswami, P., Vásquez, M., Durand, S.,
techniques for fast, convenient, and highly sensitive monitoring of im­ Wallace-Moyer, I., Zhong, L., Nett, J.H., Burnina, I., Caffry, I., Lynaugh, H.,
Sinclair, M., Sun, T., Bukowski, J., Xu, Y., Abdiche, Y.N., 2020. Assessing the binding
mune checkpoint inhibitors, such as the gated-RET, SERS, optical and properties of the anti-PD-1 antibody landscape using label-free biosensors. PLoS One
microfluidic techniques, will enable precision control over the cancer 15 (3), e0229206-e0229206.
treatment. Another method of controlling the immune response involves Cao, Y., Wang, Y., Yu, X., Jiang, X., Li, G., Zhao, J., 2020. Identification of programmed
death ligand-1 positive exosomes in breast cancer based on DNA amplification-
downregulation of tumor PD-L1 expression by gene-silencing utilizing
responsive metal-organic frameworks. Biosens. Bioelectron. 166, 112452.
siRNA. Since a pronounced expression of PD-L1 has been found on the Chen, G., Huang, A.C., Zhang, W., Zhang, G., Wu, M., Xu, W., Yu, Z., Yang, J., Wang, B.,
surface of exosomes, which were released from cancer cells, the exoso­ Sun, H., Xia, H., Man, Q., Zhong, W., Antelo, L.F., Wu, B., Xiong, X., Liu, X., Guan, L.,
mal PD-L1 level now turns out to be now a diagnosis and predictive Li, T., Liu, S., Yang, R., Lu, Y., Dong, L., McGettigan, S., Somasundaram, R.,
Radhakrishnan, R., Mills, G., Lu, Y., Kim, J., Chen, Y.H., Dong, H., Zhao, Y.,
biomarker for cancer patients’ survival rate in a noninvasive “exosomal Karakousis, G.C., Mitchell, T.C., Schuchter, L.M., Herlyn, M., Wherry, E.J., Xu, X.,
biopsy”. Although not yet applied in clinical practice, the exosomal bi­ Guo, W., 2018. Exosomal PD-L1 contributes to immunosuppression and is associated
opsy is expected to become soon one of the most important, unique, and with anti-PD-1 response. Nature 560 (7718), 382–386.
Chen, H., Wang, L., Zeng, X., Schwarz, H., Nanda, H.S., Peng, X., Zhou, Y., 2021.
robust routines in diagnostics, staging, and providing reference infor­ Exosomes, a new star for targeted delivery. Front. Cell Dev. Biol. 9.
mation for real-time monitoring and progression of cancers. There are Chou, C.-K., Huang, P.-J., Tsou, P.-H., Wei, Y., Lee, H.-H., Wang, Y.-N., Liu, Y.-L., Shi, C.,
now some commercially available products/kits with their own specific Yeh, H.-C., Kameoka, J., Hung, M.-C., 2018. A flow-proteometric platform for
analyzing protein concentration (FAP): proof of concept for quantification of PD-L1
protocols for the isolation of exosomes from bodily fluids. They can be protein in cells and tissues. Biosens. Bioelectron. 117, 97–103.
utilized for the detection and analysis of genetic or proteomic bio­ Chung, I.-M., Rajakumar, G., Venkidasamy, B., Subramanian, U., Thiruvengadam, M.,
markers (Mohammadi et al., 2021; Ohannesian et al., 2020; Lai et al., 2020. Exosomes: current use and future applications. Clin. Chim. Acta 500, 226–232.
Corthay, A., 2014. Does the immune system naturally protect against cancer? Front.
2022). Furthermore, due to a high drug-carrying capacity, non-cytotoxic Immunol. 5, 197, 197.
effects, and a low immunogenic profile of exosomes, we believe that the Couzin-Frankel, J., 2013. Cancer immunotherapy. Science 342 (6165), 1432–1433.
encapsulation of anticancer drugs and immune checkpoint inhibitors in Davis, A.A., Patel, V.G., 2019. The role of PD-L1 expression as a predictive biomarker: an
analysis of all US Food and Drug Administration (FDA) approvals of immune
exosomal nanocarriers for targeted drug delivery is likely to be one of
checkpoint inhibitors. J Immunother Cancer 7 (1), 278.
the most exciting paths leading in the near future to novel cancer de Filette, J., Andreescu, C.E., Cools, F., Bravenboer, B., Velkeniers, B., 2019.
therapies aiming at prevention of metastasis which is the key point for A systematic review and meta-analysis of endocrine-related adverse events
improving the patients’ survivability. associated with immune checkpoint inhibitors. Horm. Metab. Res. 51 (3), 145–156.
Dong, H., Zhu, G., Tamada, K., Chen, L., 1999. B7-H1, a third member of the B7 family,
co-stimulates T-cell proliferation and interleukin-10 secretion. Nat. Med. 5 (12),
Declaration of competing interest 1365–1369.
Doroshow, D.B., Bhalla, S., Beasley, M.B., Sholl, L.M., Kerr, K.M., Gnjatic, S., Wistuba, I.
I., Rimm, D.L., Tsao, M.S., Hirsch, F.R., 2021. PD-L1 as a biomarker of response to
The authors declare no conflict of interest. immune-checkpoint inhibitors. Nat. Rev. Clin. Oncol. 18 (6), 345–362.
Dougall, W.C., Kurtulus, S., Smyth, M.J., Anderson, A.C., 2017. TIGIT and CD96: new
Data availability checkpoint receptor targets for cancer immunotherapy. Immunol. Rev. 276 (1),
112–120.
Farkona, S., Diamandis, E.P., Blasutig, I.M., 2016. Cancer immunotherapy: the beginning
No data was used for the research described in the article. of the end of cancer? BMC Med. 14 (1), 73.
Feng, S., Isayev, O., Werner, J., Bazhin, A.V., 2023. CD96 as a potential immune
regulator in cancers. Int. J. Mol. Sci. 24 (2).
Acknowledgments Fritz, J.M., Lenardo, M.J., 2019. Development of immune checkpoint therapy for cancer.
J. Exp. Med. 216 (6), 1244–1254.
This research was supported by funding provided by the Program García-Aranda, M., Redondo, M., 2019. Immunotherapy: a challenge of breast cancer
treatment. Cancers 11 (12), 1822.
OPUS of the National Science Centre, Poland, Grant No. 2017/25/B/ Garrett, M.D., Collins, I., 2011. Anticancer therapy with checkpoint inhibitors: what,
ST4/01362. Financial support from the Foundation for Polish Science where and when? Trends Pharmacol. Sci. 32 (5), 308–316.
(FNP) through START program (to K.R.) and Program SONATA BIS of

14
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Ghiotto, M., Gauthier, L., Serriari, N., Pastor, S., Truneh, A., Nunès, J.A., Olive, D., 2010. Ksienski, D., Wai, E.S., Croteau, N., Fiorino, L., Brooks, E., Poonja, Z., Fenton, D.,
PD-L1 and PD-L2 differ in their molecular mechanisms of interaction with PD-1. Int. Geller, G., Glick, D., Lesperance, M., 2019. Efficacy of nivolumab and
Immunol. 22 (8), 651–660. pembrolizumab in patients with advanced non-small-cell lung cancer needing
Gong, L., Yan, Q., Zhang, Y., Fang, X., Liu, B., Guan, X., 2019. Cancer cell treatment interruption because of adverse events: a retrospective multicenter
reprogramming: a promising therapy converting malignancy to benignity. Cancer analysis. Clin. Lung Cancer 20 (1), e97–e106.
Commun. 39 (1), 48. Kwak, G., Kim, D., Nam, G.-h., Wang, S.Y., Kim, I.-S., Kim, S.H., Kwon, I.-C., Yeo, Y.,
Gonzalez, H., Hagerling, C., Werb, Z., 2018. Roles of the immune system in cancer: from 2017. Programmed cell death protein ligand-1 silencing with
tumor initiation to metastatic progression. Gene Dev. 32 (19–20), 1267–1284. polyethylenimine–dermatan sulfate complex for dual inhibition of melanoma
Goodman, A., Patel, S.P., Kurzrock, R., 2017. PD-1–PD-L1 immune-checkpoint blockade growth. ACS Nano 11 (10), 10135–10146.
in B-cell lymphomas. Nat. Rev. Clin. Oncol. 14 (4), 203–220. Kwak, S.Y., Lee, S., Han, H.D., Chang, S., Kim, K.-p., Ahn, H.J., 2019. PLGA nanoparticles
Grel, H., Ratajczak, K., Jakiela, S., Stobiecka, M., 2020. Gated resonance energy transfer codelivering siRNAs against programmed cell death protein-1 and its ligand gene for
(gRET) controlled by programmed death protein ligand 1. Nanomaterials 10 (8), suppression of colon tumor growth. Mol. Pharm. 16 (12).
1592. Lai, J.J., Chau, Z.L., Chen, S.-Y., Hill, J.J., Korpany, K.V., Liang, N.-W., Lin, L.-H., Lin, Y.-
Grewal, Y.S., Shiddiky, M.J.A., Spadafora, L.J., Cangelosi, G.A., Trau, M., 2015. H., Liu, J.K., Liu, Y.-C., Lunde, R., Shen, W.-T., 2022. Exosome processing and
Structural characterization of nanoyeast single-chain fragment variable affinity characterization approaches for research and technology development. Adv. Sci. 9
reagents. J. Phys. Chem. C 119 (22), 12674–12680. (15), 2103222.
Grewal, Y.S., Shiddiky, M.J.A., Mahler, S.M., Cangelosi, G.A., Trau, M., 2016. Nanoyeast Leach, D.R., Krummel, M.F., Allison, J.P., 1996. Enhancement of antitumor immunity by
and other cell envelope compositions for protein studies and biosensor applications. CTLA-4 blockade. Science 271 (5256), 1734–1736.
ACS Appl. Mater. Interfaces 8 (45), 30649–30664. Lee, J., Lee, J.-H., Chakraborty, K., Hwang, J., Lee, Y.-K., 2022. Exosome-based drug
Gunjur, A., Manrique-Rincón, A.J., Klein, O., Behren, A., Lawley, T.D., Welsh, S.J., delivery systems and their therapeutic applications. RSC Adv. 12 (29),
Adams, D.J., 2022. ’Know thyself’ - host factors influencing cancer response to 18475–18492.
immune checkpoint inhibitors. J. Pathol. 257 (4), 513–525. Li, Y., Li, F., Jiang, F., Lv, X., Zhang, R., Lu, A., Zhang, G., 2016. A mini-review for cancer
Gyukity-Sebestyén, E., Harmati, M., Dobra, G., Németh, I.B., Mihály, J., Zvara, Á., immunotherapy: molecular understanding of PD-1/PD-L1 pathway & translational
Hunyadi-Gulyás, É., Katona, R., Nagy, I., Horváth, P., Bálind, Á., Szkalisity, Á., blockade of immune checkpoints. Int. J. Mol. Sci. 17 (7), 1151.
Kovács, M., Pankotai, T., Borsos, B., Erdélyi, M., Szegletes, Z., Veréb, Z.J., Buzás, E.I., Li, J., Wang, J., Grewal, Y.S., Howard, C.B., Raftery, L.J., Mahler, S., Wang, Y., Trau, M.,
Kemény, L., Bíró, T., Buzás, K., 2019. Melanoma-derived exosomes induce PD-1 2018. Multiplexed SERS detection of soluble cancer protein biomarkers with
overexpression and tumor progression via mesenchymal stem cell oncogenic gold–silver alloy nanoboxes and nanoyeast single-chain variable fragments. Anal.
reprogramming. Front. Immunol. 10. Chem. 90 (17), 10377–10384.
Han, Y., Gao, Y., He, T., Wang, D., Guo, N., Zhang, X., Chen, S., Wang, H., 2018. PD-1/ Li, C., Li, C., Zhi, C., Liang, W., Wang, X., Chen, X., Lv, T., Shen, Q., Song, Y., Lin, D.,
PD-L1 inhibitor screening of caffeoylquinic acid compounds using surface plasmon Liu, H., 2019a. Clinical significance of PD-L1 expression in serum-derived exosomes
resonance spectroscopy. Anal. Biochem. 547, 52–56. in NSCLC patients. J. Transl. Med. 17 (1), 355.
Han, Y., Liu, D., Li, L., 2020. PD-1/PD-L1 pathway: current researches in cancer. Am. J. Li, G., Gao, Y., Gong, C., Han, Z., Qiang, L., Tai, Z., Tian, J., Gao, S., 2019b. Dual-
Cancer Res. 10 (3), 727–742. blockade immune checkpoint for breast cancer treatment based on a tumor-
Harjunpää, H., Guillerey, C., 2019. TIGIT as an emerging immune checkpoint. Clin. Exp. penetrating peptide assembling nanoparticle. ACS Appl. Mater. Interfaces 11 (43),
Immunol. 200 (2), 108–119. 39513–39524.
Homet Moreno, B., Ribas, A., 2015. Anti-programmed cell death protein-1/ligand-1 Li, M., Li, S., Du, C., Zhang, Y., Li, Y., Chu, L., Han, X., Galons, H., Zhang, Y., Sun, H.,
therapy in different cancers. Br. J. Cancer 112 (9), 1421–1427. Yu, P., 2020. Exosomes from different cells: characteristics, modifications, and
Hu, J., Zhang, Z.-h., Zhu, Z., Chen, J., Hu, X., Chen, H., 2021. Specific intracellular therapeutic applications. Eur. J. Med. Chem. 207, 112784.
binding peptide as sPD-L1 antibody mimic: robust binding capacity and intracellular Lipson, E.J., Forde, P.M., Hammers, H.-J., Emens, L.A., Taube, J.M., Topalian, S.L., 2015.
region specific modulation upon applied to sensing research. Biosens. Bioelectron. Antagonists of PD-1 and PD-L1 in cancer treatment. Semin. Oncol. 42 (4), 587–600.
185, 113269. Liu, A., Dong, L., Wei, X.-L., Yang, X.-H., Xiao, J.-H., Liu, Z.-Q., 2016. Development of
Hu-Lieskovan, S., Ribas, A., 2017. New combination strategies using programmed cell amino- and dimethylcarbamate-substituted resorcinol as programmed cell death-1
death 1/programmed cell death ligand 1 checkpoint inhibitors as a backbone. (PD-1) inhibitor. Eur. J. Pharmaceut. Sci. 88, 50–58.
Cancer J. 23 (1), 10–22. Liu, C., Zeng, X., An, Z., Yang, Y., Eisenbaum, M., Gu, X., Jornet, J.M., Dy, G.K., Reid, M.
Huang, P.-W., Chang, J.W.-C., 2019. Immune checkpoint inhibitors win the 2018 Nobel E., Gan, Q., Wu, Y., 2018. Sensitive detection of exosomal proteins via a compact
Prize. Biomed. J. 42 (5), 299–306. surface plasmon resonance biosensor for cancer diagnosis. ACS Sens. 3 (8),
Huang, X., Zhang, Z.-h., Chen, J., Mao, Z., Zhu, H., Liu, Y., Zhu, Z., Chen, H., 2021. One 1471–1479.
dimensional magneto-optical nanocomplex from silver nanoclusters and magnetite Liu, H., Zhao, Z., Zhang, L., Li, Y., Jain, A., Barve, A., Jin, W., Liu, Y., Fetse, J., Cheng, K.,
nanorods containing ordered mesocages for sensitive detection of PD-L1. Biosens. 2019. Discovery of low-molecular weight anti-PD-L1 peptides for cancer
Bioelectron. 189, 113385. immunotherapy. J Immunother Cancer 7 (1), 270.
Hung, A.L., Maxwell, R., Theodros, D., Belcaid, Z., Mathios, D., Luksik, A.S., Kim, E., Liu, F., Huang, J., He, F., Ma, X., Fan, F., Meng, M., Zhuo, Y., Zhang, L., 2020. CD96, a
Wu, A., Xia, Y., Garzon-Muvdi, T., Jackson, C., Ye, X., Tyler, B., Selby, M., new immune checkpoint, correlates with immune profile and clinical outcome of
Korman, A., Barnhart, B., Park, S.-M., Youn, J.-I., Chowdhury, T., Park, C.-K., glioma. Sci. Rep. 10 (1), 10768.
Brem, H., Pardoll, D.M., Lim, M., 2018. TIGIT and PD-1 dual checkpoint blockade Liu, C., Seeram, N.P., Ma, H., 2021. Small molecule inhibitors against PD-1/PD-L1
enhances antitumor immunity and survival in GBM. OncoImmunology 7 (8), immune checkpoints and current methodologies for their development: a review.
e1466769. Cancer Cell Int. 21 (1), 239.
Incorvaia, L., Fanale, D., Badalamenti, G., Barraco, N., Bono, M., Corsini, L.R., Liu, J., Chen, Z., Li, Y., Zhao, W., Wu, J., Zhang, Z., 2021. PD-1/PD-L1 checkpoint
Galvano, A., Gristina, V., Listì, A., Vieni, S., Gori, S., Bazan, V., Russo, A., 2019. inhibitors in tumor immunotherapy. Front. Pharmacol. 12, 731798.
Programmed death ligand 1 (PD-L1) as a predictive biomarker for pembrolizumab Luan, Y., Chai, D., Peng, J., Ma, S., Wang, M., Ma, H., Li, X., Fu, S., Pan, X., Wang, X.,
therapy in patients with advanced non-small-cell lung cancer (NSCLC). Adv. Ther. 36 Qin, S., Xu, T., 2016. A fully human monoclonal antibody targeting PD-L1 with
(10), 2600–2617. potent anti-tumor activity. Int. Immunopharm. 31, 248–256.
Ishida, Y., Agata, Y., Shibahara, K., Honjo, T., 1992. Induced expression of PD-1, a novel Luo, B., Wang, Y., Lu, H., Wu, S., Lu, Y., Shi, S., Li, L., Jiang, S., Zhao, M., 2019. Label-
member of the immunoglobulin gene superfamily, upon programmed cell death. free and specific detection of soluble programmed death ligand-1 using a localized
EMBO J. 11 (11), 3887–3895. surface plasmon resonance biosensor based on excessively tilted fiber gratings.
Iwai, Y., Ishida, M., Tanaka, Y., Okazaki, T., Honjo, T., Minato, N., 2002. Involvement of Biomed. Opt Express 10 (10), 5136–5148.
PD-L1 on tumor cells in the escape from host immune system and tumor Lux, A., Kahlert, C., Grützmann, R., Pilarsky, C., 2019. c-Met and PD-L1 on circulating
immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. U. S. A. 99 (19), exosomes as diagnostic and prognostic markers for pancreatic cancer. Int. J. Mol. Sci.
12293–12297. 20 (13), 3305.
Janssen, L.M.E., Ramsay, E.E., Logsdon, C.D., Overwijk, W.W., 2017. The immune Macek Jilkova, Z., Aspord, C., Decaens, T., 2019. Predictive factors for response to PD-1/
system in cancer metastasis: friend or foe? J Immunother Cancer 5 (1), 79. PD-L1 checkpoint inhibition in the field of hepatocellular carcinoma: current status
Kar, R., Dhar, R., Mukherjee, S., Nag, S., Gorai, S., Mukerjee, N., Mukherjee, D., Vatsa, R., and challenges. Cancers 11 (10), 1554.
Chandrakanth Jadhav, M., Ghosh, A., Devi, A., Krishnan, A., Thorat, N.D., 2023. Mahoney, K.M., Freeman, G.J., McDermott, D.F., 2015. The next immune-checkpoint
Exosome-based smart drug delivery tool for cancer theranostics. ACS Biomater. Sci. inhibitors: PD-1/PD-L1 blockade in melanoma. Clin. Therapeut. 37 (4), 764–782.
Eng. 9 (2), 577–594. Makker, V., Taylor, M.H., Aghajanian, C., Oaknin, A., Mier, J., Cohn, A.L., Romeo, M.,
Kaufman, H.L., Atkins, M.B., Subedi, P., Wu, J., Chambers, J., Mattingly II, T.J., Bratos, R., Brose, M.S., DiSimone, C., Messing, M., Stepan, D.E., Dutcus, C.E., Wu, J.,
Campbell, J.D., Allen, J., Ferris, A.E., Schilsky, R.L., Danielson, D., Lichtenfeld, J.L., Schmidt, E.V., Orlowski, R., Sachdev, P., Shumaker, R., Casado Herraez, A., 2020.
House, L., Selig, W.K.D., 2019. The promise of Immuno-oncology: implications for Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer.
defining the value of cancer treatment. J Immunother Cancer 7 (1), 129. J. Clin. Oncol. : official journal of the American Society of Clinical Oncology 38 (26),
Kim, D.H., Kim, H.R., Choi, Y.J., Kim, S.Y., Lee, J.-E., Sung, K.J., Sung, Y.H., Pack, C.-G., 2981–2992.
Jung, M.-k., Han, B., Kim, K., Kim, W.S., Nam, S.J., Choi, C.-M., Yun, M., Lee, J.C., Mariam, A., Kamath, S., Schveder, K., McLeod, H.L., Rotroff, D.M., 2023. Biomarkers for
Rho, J.K., 2019. Exosomal PD-L1 promotes tumor growth through immune escape in response to anti-PD-1/anti-PD-L1 immune checkpoint inhibitors: a large meta-
non-small cell lung cancer. Exp. Mol. Med. 51 (8), 94. analysis. Oncology 37 (5), 210–219.
Kosmides, A.K., Sidhom, J.-W., Fraser, A., Bessell, C.A., Schneck, J.P., 2017. Dual Marshall, H.T., Djamgoz, M.B.A., 2018. Immuno-Oncology: emerging targets and
targeting nanoparticle stimulates the immune system to inhibit tumor growth. ACS combination therapies. Front. Oncol. 8 (315).
Nano 11 (6), 5417–5429. Mittal, D., Lepletier, A., Madore, J., Aguilera, A.R., Stannard, K., Blake, S.J.,
Whitehall, V.L.J., Liu, C., Bettington, M.L., Takeda, K., Long, G.V., Scolyer, R.A.,

15
K. Ratajczak et al. Biosensors and Bioelectronics 240 (2023) 115644

Lan, R., Siemers, N., Korman, A., Teng, M.W.L., Johnston, R.J., Dougall, W.C., Stobiecka, M., Chalupa, A., 2015. Modulation of plasmon-enhanced resonance energy
Smyth, M.J., 2019. CD96 is an immune checkpoint that regulates CD8(+) T-cell transfer to gold nanoparticles by protein survivin channeled-shell gating. J. Phys.
antitumor function. Cancer Immunol. Res. 7 (4), 559–571. Chem. B 119, 13227–13235.
Mohammadi, M., Zargartalebi, H., Salahandish, R., Aburashed, R., Wey Yong, K., Sanati- Tan, S., Liu, K., Chai, Y., Zhang, C.W.H., Gao, S., Gao, G.F., Qi, J., 2018. Distinct PD-L1
Nezhad, A., 2021. Emerging technologies and commercial products in exosome- binding characteristics of therapeutic monoclonal antibody durvalumab. Protein &
based cancer diagnosis and prognosis. Biosens. Bioelectron. 183, 113176. Cell 9 (1), 135–139.
Morrissey, S.M., Yan, J., 2020. Exosomal PD-L1: roles in tumor progression and Tang, Y., Zhang, P., Wang, Y., Wang, J., Su, M., Wang, Y., Zhou, L., Zhou, J., Xiong, W.,
immunotherapy. Trends in Cancer 6 (7), 550–558. Zeng, Z., Zhou, Y., Nie, S., Liao, Q., 2020. The biogenesis, biology, and clinical
Mu, X., Zhang, M., Wei, A., Yin, F., Wang, Y., Hu, K., Jiang, J., 2021. Doxorubicin and significance of exosomal PD-L1 in cancer. Front. Immunol. 11, 604, 604.
PD-L1 siRNA co-delivery with stem cell membrane-coated polydopamine Theodoraki, M.-N., Yerneni, S.S., Hoffmann, T.K., Gooding, W.E., Whiteside, T.L., 2018.
nanoparticles for the targeted chemoimmunotherapy of PCa bone metastases. Clinical significance of PD-L1(+) exosomes in plasma of head and neck cancer
Nanoscale 13 (19), 8998–9008. patients. Clin. Cancer Res. 24 (4), 896–905.
Niu, J., Maurice-Dror, C., Lee, D.H., Kim, D.W., Nagrial, A., Voskoboynik, M., Chung, H. Thibaudin, M., Limagne, E., Hampe, L., Ballot, E., Truntzer, C., Ghiringhelli, F., 2022.
C., Mileham, K., Vaishampayan, U., Rasco, D., Golan, T., Bauer, T.M., Jimeno, A., Targeting PD-L1 and TIGIT could restore intratumoral CD8 T cell function in human
Chung, V., Chartash, E., Lala, M., Chen, Q., Healy, J.A., Ahn, M.J., 2022. First-in- colorectal cancer. Cancer Immunol. Immunother. 71 (10), 2549–2563.
human phase 1 study of the anti-TIGIT antibody vibostolimab as monotherapy or Villasboas, J.C., Ansell, S., 2016. Checkpoint inhibition: programmed cell death 1 and
with pembrolizumab for advanced solid tumors, including non-small-cell lung programmed cell death 1 ligand inhibitors in Hodgkin lymphoma. Cancer J. 22 (1),
cancer(☆). Ann. Oncol. 33 (2), 169–180. 17–22.
Novotny Jr., J.F., Cogswell, J., Inzunza, H., Harbison, C., Horak, C., Averbuch, S., 2016. Wang, Y., Wang, C., Qiu, J., Qu, X., Peng, J., Lu, C., Zhang, M., Zhang, M., Qi, X., Li, G.,
Establishing a complementary diagnostic for anti-PD-1 immune checkpoint inhibitor Hua, K., 2022. Targeting CD96 overcomes PD-1 blockade resistance by enhancing
therapy. Ann. Oncol. 27 (10), 1966–1969. CD8+ TIL function in cervical cancer. Journal for ImmunoTherapy of Cancer 10 (3),
Ohannesian, N., Gunawardhana, L., Misbah, I., Rakhshandehroo, M., Lin, S.H., Shih, W.- e003667.
C., 2020. Commercial and emerging technologies for cancer diagnosis and prognosis Wang, M., Wisniewski, C.A., Xiong, C., Chhoy, P., Goel, H.L., Kumar, A., Zhu, L.J., Li, R.,
based on circulating tumor exosomes. J. Phys.: Photonics 2, 032002. St Louis, P.A., Ferreira, L.M., Pakula, H., Xu, Z., Loda, M., Jiang, Z., Brehm, M.A.,
Pandya, P.H., Murray, M.E., Pollok, K.E., Renbarger, J.L., 2016. The immune system in Mercurio, A.M., 2023. Therapeutic blocking of VEGF binding to neuropilin-2
cancer pathogenesis: potential therapeutic approaches. J Immunol Res 2016, diminishes PD-L1 expression to activate antitumor immunity in prostate cancer. Sci.
4273943. Transl. Med. 15 (694), eade5855.
Pang, Y., Shi, J., Yang, X., Wang, C., Sun, Z., Xiao, R., 2020. Personalized detection of https://www.webofscience.com/wos/woscc/citation-report/5dddc8d6-e2f9-4c2b-a7a2
circling exosomal PD-L1 based on Fe3O4@TiO2 isolation and SERS immunoassay. -62e9a7416f54-959ad3d6, 2023. www.webofknowledge.com. In: Science, W.o.
Biosens. Bioelectron. 148, 111800. (Ed.), p. Clarivate Analytics. Clarivate Analytics.
Pardoll, D.M., 2012. The blockade of immune checkpoints in cancer immunotherapy. Weigmann, K., 2016. Releasing the brakes to fight cancer: the recent discovery of
Nat. Rev. Cancer 12 (4), 252–264. checkpoints has boosted the field of cancer immunotherapy. EMBO Rep. 17 (9),
Passiglia, F., Bronte, G., Bazan, V., Natoli, C., Rizzo, S., Galvano, A., Listì, A., Cicero, G., 1257–1260.
Rolfo, C., Santini, D., Russo, A., 2016. PD-L1 expression as predictive biomarker in 2020 WHO report on cancer: setting priorities, investing wisely and providing care for
patients with NSCLC: a pooled analysis. Oncotarget 7 (15). all. In: Organization, W.H. (Ed.). World Health Organization.
Patel, S.P., Kurzrock, R., 2015. PD-L1 expression as a predictive biomarker in cancer Wu, X., Gu, Z., Chen, Y., Chen, B., Chen, W., Weng, L., Liu, X., 2019a. Application of PD-1
immunotherapy. Mol. Cancer Therapeut. 14 (4), 847–856. blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 17, 661–674.
Patel, A.J., Willsmore, Z.N., Khan, N., Richter, A., Naidu, B., Drayson, M.T., Papa, S., Wu, Y., Chen, W., Xu, Z.P., Gu, W., 2019b. PD-L1 distribution and perspective for cancer
Cope, A., Karagiannis, S.N., Perucha, E., Middleton, G.W., 2022. Regulatory B cell immunotherapy-blockade, knockdown, or inhibition. Front. Immunol. 10, 2022,
repertoire defects predispose lung cancer patients to immune-related toxicity 2022.
following checkpoint blockade. Nat. Commun. 13 (1), 3148. Wuethrich, A., Rajkumar, A.R., Shanmugasundaram, K.B., Reza, K.K., Dey, S.,
Philips, G.K., Atkins, M., 2014. Therapeutic uses of anti-PD-1 and anti-PD-L1 antibodies. Howard, C.B., Sina, A.A.I., Trau, M., 2019. Single droplet detection of immune
Int. Immunol. 27 (1), 39–46. checkpoints on a multiplexed electrohydrodynamic biosensor. Analyst 144 (23),
Poggio, M., Hu, T., Pai, C.-C., Chu, B., Belair, C.D., Chang, A., Montabana, E., Lang, U.E., 6914–6921.
Fu, Q., Fong, L., Blelloch, R., 2019. Suppression of exosomal PD-L1 induces systemic Xia, A.-L., Xu, Y., Lu, X.-J., 2019. Cancer immunotherapy: challenges and clinical
anti-tumor immunity and memory. Cell 177 (2), 414–427.e413. applications. J. Med. Genet. 56 (1), 1–3.
Reza, K.K., Sina, A., Wuethrich, A., Grewal, Y.S., Howard, C.B., Korbie, D., Trau, M., Xie, Y.-Q., Wei, L., Tang, L., 2018. Immunoengineering with biomaterials for enhanced
2019. A SERS microfluidic platform for targeting multiple soluble immune cancer immunotherapy. Wires Nanomed Nanobi 10 (4), e1506.
checkpoints. Biosens. Bioelectron. 126, 178–186. Xie, F., Xu, M., Lu, J., Mao, L., Wang, S., 2019. The role of exosomal PD-L1 in tumor
Ribas, A., 2012. Tumor immunotherapy directed at PD-1. N. Engl. J. Med. 366, progression and immunotherapy. Mol. Cancer 18 (1), 146.
2517–2519. Xu, F., Jin, T., Zhu, Y., Dai, C., 2018. Immune checkpoint therapy in liver cancer. J. Exp.
Riley, R.S., June, C.H., Langer, R., Mitchell, M.J., 2019. Delivery technologies for cancer Clin. Cancer Res. 37 (1), 110.
immunotherapy. Nat. Rev. Drug Discov. 18 (3), 175–196. Yazdian-Robati, R., Ramezani, M., Khedri, M., Ansari, N., Abnous, K., Taghdisi, S.M.,
Santarpia, M., Karachaliou, N., 2015. Tumor immune microenvironment 2017. An aptamer for recognizing the transmembrane protein PDL-1 (programmed
characterization and response to anti-PD-1 therapy. Cancer Biol Med 12 (2), 74–78. death-ligand 1), and its application to fluorometric single cell detection of human
Sen, S., Xavier, J., Kumar, N., Ahmad, M.Z., Ranjan, O.P., 2023. Exosomes as natural ovarian carcinoma cells. Microchim. Acta 184 (10), 4029–4035.
nanocarrier-based drug delivery system: recent insights and future perspectives. 3 Zak, K.M., Kitel, R., Przetocka, S., Golik, P., Guzik, K., Musielak, B., Dömling, A.,
Biotech 13 (3), 101. Dubin, G., Holak, T.A., 2015. Structure of the complex of human programmed death
Siegel, R.L., Miller, K.D., Fuchs, H.E., Jemal, A., 2021. Cancer statistics, 2021. CA A 1, PD-1, and its ligand PD-L1. Structure 23 (12), 2341–2348.
Cancer J. Clin. 71 (1), 7–33. Zang, X., 2018. 2018 Nobel Prize in medicine awarded to cancer immunotherapy:
Song, Y., Wu, L., Yang, C., 2019. Exosomal PD-L1: an effective liquid biopsy target to immune checkpoint blockade - a personal account. Genes Dis 5 (4), 302–303.
predict immunotherapy response. Natl. Sci. Rev. 6, 1103–1104. Zhang, H., Wang, S., Sun, M., Cui, Y., Xing, J., Teng, L., Xi, Z., Yang, Z., 2023a. Exosomes
Stobiecka, M., 2014. Novel plasmonic field-enhanced nanoassay for trace detection of as smart drug delivery vehicles for cancer immunotherapy. Front. Immunol. 13.
proteins. Biosens. Bioelectron. 55, 379–385. Zhang, M., Hu, S., Liu, L., Dang, P., Liu, Y., Sun, Z., Qiao, B., Wang, C., 2023b.
Stobiecka, M., 2015. Exosomes and microvesicles: extracellular frontiers of intercellular Engineered exosomes from different sources for cancer-targeted therapy. Signal
communication. In: Somasundaran, P. (Ed.), Encyclopedia of Surface and Colloid Transduct. Targeted Ther. 8 (1), 124.
Science, third ed. CRC Press Taylor & Francis Group, New York. Zhao, J., Li, L., Yin, H., Feng, X., Lu, Q., 2023. TIGIT: an emerging immune checkpoint
target for immunotherapy in autoimmune disease and cancer. Int. Immunopharm.
120, 110358.

16

You might also like