Prés Molecular Imaging - HES

publicité
Healthcare economics
Shift of paradigm
Transformation of medical paradigm
New developments in
Molecular
Imaging
•  Détecteur
à scintillations
Applications in personalized medicine
spectromètre
Osman Ratib, MD, PhD
• Treat disease before it becomes symptomatic
• Four P
PM
cristal
collimateur
«name» - «studyName»
Molecular Imaging
Evolution of medical imaging
Predictive
Personalized
Preemptive
Participatory
• We should better understand how the
molecules we are giving to the patients really
work (how cancer mutate, response,
environment etc..)
Division of Nuclear Medicine and Molecular Imaging
Department of Medical Imaging and Information Sciences
University Hospital of Geneva
Image 1
•
•
•
•
Keynote Lecture
Elias Zerhouni
International Society
for Strategic Studies
in Radiology
•
•
•
Genomics, proteomics
Molecular Imaging (quantitative…)
Radiomics
©2016 Hôpitaux Universitaires de Genève
asservissement
mécanique
imprimante
HISTORY
Evolution of molecular imaging
1958
2015
Planar scintigram
Hybrid PET-CT
➩ scintigraphie
©2011 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Master Physique Médicale Lyon - Tomographie d’émission monophotonique - Irène Buvat – septembre 2011 - 26
Evolution of medical imaging
Molecular Imaging
Multidetector scanners
The fifth dimension
3D
4D
©2014 Hôpitaux Universitaires de Genève
Positron emission tomography
Molecular imaging
Anatomy (CT)
Metabolism (PET)
©2014 Hôpitaux Universitaires de Genève
PET vs CT
Advantages
CT
Pre treatment
Post treatment
©2014 Hôpitaux Universitaires de Genève
©2011 Hôpitaux Universitaires de Genève
PET
Positron Emission Tomography
Evolution of hybrid Imaging
Invention of hybrid PET-CT
PET-CT scanners
1994
1998
University of Pittsburgh
Medical Center
©2006 Hôpitaux Universitaires de Genève
©2008 Hôpitaux Universitaires de Genève
PET-CT
PET-CT
Staging in oncology
Treatment monitoring
Follow up of treatment response
Better characterization of lesions
T.M. 3487591
©2011 Hôpitaux Universitaires
de Genève
Staging
Post therapy
T.M. 3487591
©2011 Hôpitaux Universitaires
de Genève
Staging
Mobile PET-CT
Hybrid imaging
From PET-CT to PET-MR
CT
PET
PET-CT
PET-MRI
CT
PET
PET-CT
©2009 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Hybrid PET-MRI
Evolution...
Evolution...
PET
©2016 Hôpitaux Universitaires de Genève
MRI
PET-MRI
Hybrid PET-MRI
Separate
MRI
PET
MRI
Separate
©2016 Hôpitaux Universitaires de Genève
➟ Co-planar
MRI
Hybrid PET-MRI
Evolution...
PET-MR
hybrid imaging
PET
MRI
World premier in hybrid imaging...
Separate
➟ Co-planar
➟ Integrated
©2016 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Hybrid whole-body PET-MRI
Hybrid PET-MR
Co-planar registration of standard scanners
Clinical applications of hybrid PET-MR
• Le PET-CT est un nouvel outil diagnostic pour la détection et
le suivi des cancers
3m
MRI
• L’acquisition simultanée de CT et de PET offre des avantages
de qualité diagnostique et de confort pour le patient
• Le PET offre aussi des applications en neurologie, cardiologie
et dans la détection des infections et des foyers
inflammatoires
PET
• Le développement de nouveaux traceurs plus spécifiques
ouvre de nouvelles perspectives dans le diagnostique
précoce des tumeurs et dans l’étude des maladies
neurodégénératives et cardiovasculaires
©2016 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Hybrid PET-MR
Clinical applications of hybrid PET-MR
Head & Neck
Breast
Prostate
Sarcoma
Pelvis
from diagnostic to
Theranostic
A new paradigm in molecular imaging...
PET$CT
©2016 Hôpitaux Universitaires de Genève
THERANOSTICS FOR PERSONALIZED
MOLECULAR TARGETED THERAPY
Shift of paradigm
©2016 Hôpitaux Universitaires de Genève
HISTORY
Radionuclide can cure cancer..
Theranostics
• Theranostics is the combination of a Diagnostic Tool that also
provides a Therapeutic Tool for a specific disease.
• The right treatment, not anymore targeting the “specific disease”
but the “specific disease expression of a given patient”.
Personalized Medicine
• The right treatment, for the right patient, at the right time, at the
right dose. – first time
• The concept of PM has been extended to Personalized Health Care
that includes all steps from the first sign of disease up to full
recovery
Published: May 12th 1921
© The New York Times
©2016 Hôpitaux Universitaires de Genève
©2015 Hôpitaux Universitaires de Genève
Targeted Radionuclide Therapy
Targeted Radionuclide Therapy
Radium-223 (Alpharadin)
Radium-223 (Alpharadin)
•
1921
223
Ra (Alpharadin) for treatment of bone metastases in
patients with prostate cancer resistant to hormonotherapy
2015
©2015 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Targeted Radionuclide Therapy
Targeted Radionuclide Therapy
Radium-223 (Alpharadin)
Radium-223 (Alpharadin)
• Phase II and phase III studies showed
increase in overall survival rate of 3.5
months
• Increase in time to first skeletal-related
event of 5.5 months
©2016 Hôpitaux Universitaires de Genève
Nilsson et al. Lancet Oncol 2007
©2016 Hôpitaux Universitaires de Genève
HISTORY
Radionuclide therapy
Theranostics in nuclear medicine
131
I Therapy for thyroid cancer
Eur J Nucl Med Mol Imaging (2014) 41:199–201
DOI 10.1007/s00259-013-2609-2
EDITORIAL
Nothing new under the nuclear sun: towards 80 years
of theranostics in nuclear medicine
• Well established since the 1950s
• Beta and gamma rays
• Self-targeting
Frederik A. Verburg & Alexander Heinzel &
Heribert Hänscheid & Felix M. Mottaghy &
Markus Luster & Luca Giovanella
Published online: 6 November 2013
# Springer-Verlag Berlin Heidelberg 2013
Some time in the early 2000s, the word “theranostics” (or
“theragnostics”) started surfacing in the medical literature.
Theranostics (from the Greek therapeuein “to treat medically”
and gnosis “knowledge”) is the use of individual patient-level
biological information in choosing the optimal therapy for that
individual [1]. In the modern era of “personalized medicine”,
theranostics is increasingly pursued in many branches of
medicine in order to develop ever more effective treatment
regimens. There are now many studies and reviews dedicated
to theranostics, and even a journal bearing the name of this
principle, detailing many different concepts on how to
combine imaging and therapy using, for example, complex
molecules [2] or nanotechnology [3].
However, it is rarely realized by either clinicians or scientists
©2016 Hôpitaux Universitaires
de Genève
that nuclear
medicine has been employing theranostics for
nearly 80 years now. In fact, the very foundations of targeted
therapy in nuclear medicine are those that are only now being
adopted by other medical disciplines under the designation
“theranostics”.
F. A. Verburg (*) : A. Heinzel : F. M. Mottaghy
Department of Nuclear Medicine, RWTH University Hospital
Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
e-mail: [email protected]
The cornerstones of theranostics can be traced back to some
of the most illustrious names among the founding fathers of
nuclear medicine. Soon after Chiewitz and de Hevesy [4]
described the uptake of radioactive 32P in the bones of rats, Erf
and J.H. Lawrence (brother of the physicist Ernest O. Lawrence,
who built the first cyclotron) applied this same radioisotope to
patients suffering from leukaemia and polycythaemia vera [5].
Although this treatment certainly was not without success, it has
since been superseded by more effective nonradioactive
chemotherapy. Shortly afterwards Pecher [6] discovered that
89
Sr accumulated in secondary bone tumours in animals, and
subsequently successfully used this radioisotope to treat patients
with painful bone metastases (unfortunately this work was
immediately classified as secret and it took more than five
decades for 89Sr to be registered as a therapeutic drug). These
two studies are perhaps the earliest examples of diagnostic
studies leading to targeted therapy of cancer using radionuclides.
Around the same time the most prominent example of pure
nuclear theranostic medicine emerged: the diagnosis and
treatment of thyroid disorders using various isotopes of iodine.
Hertz et al. in 1938 described the first study of thyroidal
radioiodine uptake [7], and in 1942 Hertz and Roberts reported
on the treatment of the first patients with Graves’ disease with
radioiodine [8]. A short time later Seidlin et al. treated the first
patient with metastatic thyroid cancer with radioiodine [9] – at
the time this compound was so rare that radioiodine was purified
from the patient’s urine and readministered. During this therapy,
additional metastases were identified using a Geiger counter and
the first rudimentary dosimetry was performed. It is of course
only with the benefit of hindsight that we can now say that this
was the first application of theranostics in targeted molecular
medicine through a specific molecular target, the sodium iodine
symporter, long before any of these concepts were first
described as “theranostics”. Indeed, even today it is hard to
think of a single combination of targeted diagnostics and therapy
that is more specific than radioiodine.
©2016 Hôpitaux Universitaires de Genève
Radionuclide therapy
Radionuclide therapy
Choice of carriers
131
F. A. Verburg : F. M. Mottaghy
Department of Nuclear Medicine, Maastricht University Medical
Center, Maastricht, The Netherlands
H. Hänscheid
Department of Nuclear Medicine, University of Wuerzburg,
Wuerzburg, Germany
M. Luster
Department of Nuclear Medicine, University Hospitals
Giessen-Marburg, Marburg, Germany
L. Giovanella
Department of Nuclear Medicine, Oncology Institute of Southern
Switzerland, Bellinzona, Switzerland
I/90Y labelled antibody for NHL therapy
• Monoclonal antibody
• Labelled with Beta emitting isotope
CD20
©2016 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Rituximab
Radionuclide therapy
131
Radionuclide therapy
I/90Y labelled antibody for NHL therapy
Treatment of indolent lymphoma with
131
Radio-immunotherapy
90
Y-Ibritumomab (Zavalin®)
I-tositumomab
Cellule B
antigène CD20
(cellule cancéreuse)
Anticorps monoclonal
(ibritumomab),
associé à un radioisotope
(yttrium-90)
L’yttrium-90 se décompose en
zirconium-90 stable, par
l’émission des particules bêta
riches en énergie (demi-vie:
2.67 jours).
La portée des rayons bêta de
l’yttrium-90 dans le tissu est de
5 mm au maximum.
F. Buchegger et al
British Journal of Cancer (2006) 94, 1770 – 1776 et J Nucl Med 2011; 52:896–900
©2016 Hôpitaux Universitaires de Genève
4
©2016 Hôpitaux Universitaires de Genève
Peptide Receptor Radiotherapy (PRRT)
68
68
PET tracers without a cyclotron
Ga Generators and labeling kits
GA/177LU DOTATOC in Neuroendocrine tumors (NET)
177
68 LU
GA
•
177
LU emits intermediate energy beta
particles (133 KeV)
• Beneficial in small sized tumors (tissue
range 2mm)
• Concomitant gamma emission enables
imaging with gamma cameras
©2016 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Radionuclide therapy
Radionuclide therapy
Peptide Receptor Radionuclide Therapy (PRRT)
Peptide Receptor Radionuclide Therapy (PRRT)
Lutathera®
Lutathera®
• Phase II results in progressive midgut
carcinoid showed Progression-Free
Survival of more than 44 months
compared to the reported 14.6 months of
Novartis' Sandostatin® LAR
• Phase II results in progressive midgut
carcinoid showed Progression-Free
Survival of more than 44 months
compared to the reported 14.6 months of
Novartis' Sandostatin® LAR
• Lutathera® was shown to increase overall
survival by between 3.5 and 6 years in
comparison to current treatments,
including chemotherapy.
• Lutathera® was shown to increase overall
survival by between 3.5 and 6 years in
comparison to current treatments,
including chemotherapy.
• It was also shown to significantly improve
quality of life
• It was also shown to significantly improve
quality of life
©2015 Hôpitaux Universitaires de Genève
©2015 Hôpitaux Universitaires de Genève
PSMA ligand
68
Prostate-Specific Membrane Antigen
Prostate-Specific Membrane Antigen
• PSMA (Prostate Specific Membrane
Antigen) is a membrane glycoprotein
which is overexpressed on prostate
cancers
• PSMA expression increases with tumor
aggressiveness, androgenindependence, metastatic disease,
and disease recurrence.
Ga labelled PSMA ligand
•
•
•
•
Type II membrane bound glycoprotein
Expressed in all forms of prostate tissue
Over-expressed in carcinoma
Also found in the neovasculature of most
solid tumors
• Ga-68 PSMA PET/CT Imaging
identifies tumor cells expressing PSMA
antigen with excellent sensitivity &
specificity.
©2015 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
68
68
Ga labelled PSMA ligand
68
Ga(HBED-CC)
18
F-Fluorocholine
Ga labelled PSMA ligand
Glu-NH-CO-NH-Lys-(Ahx)[68Ga(HBED-CC)] Compared to
18F-Fluorocholine
©2016 Hôpitaux Universitaires de Genève
Ga labelled PSMA ligand
©2016 Hôpitaux Universitaires de Genève
177Lu-labeled
F-Fluorocholine
PSMA-Targeted Radionuclide Therapy of
Prostate Cancer
PSMA-Targeted Radionuclide Therapy of
Prostate Cancer
Anti-PSMA Antibody
©2016 Hôpitaux Universitaires de Genève
PSMA-Targeted Radionuclide Therapy of
Prostate Cancer
©2016 Hôpitaux Universitaires de Genève
18
©2016 Hôpitaux Universitaires de Genève
PSMA-Targeted Radionuclide Therapy of
Prostate Cancer
Bone scan
Ga(HBED-CC)
©2016 Hôpitaux Universitaires de Genève
68
99mTC
68
99mTC
Bone scan
177Lu-labeled
Anti-PSMA Antibody
©2016 Hôpitaux Universitaires de Genève
PSMA-Targeted Radionuclide Therapy of
Prostate Cancer
©2016 Hôpitaux Universitaires de Genève
Future of theranostics
Future of theranostics
18
18
F-Galacto-RGD (Integrin ∝Vß3 receptors)
F-Galacto-RGD (Integrin ∝Vß3 receptors)
18
•
Neoangiogenesis markers ( F-Galacto RGD)
•
The ∝Vß3 integrin is up-regulated during
wound healing and cancer
•
Imaging with 18F-galacto-RGD PET has been
used for antiangiogenic therapy planning and
monitoring
∝
∝
©2016 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Future of theranostics
Future of theranostics
18
68
F-Galacto-RGD (Integrin ∝Vß3 receptors)
GA-Bombesin receptor antagonist
LEENAARDS((award((2014
A theranostic approach towards targeted chemotherapy
delivery by RGD-conjugated cytotoxic compounds
•
68
•
GRPr proteins are highly overexpressed
in several human tumors, including
prostate cancer
Dr Anita Wolfer (CHUV), Pr. Elena Dubikovskaya (EPFL), Pr. Yann Seimbille (UniGE-HUG)
©2016 Hôpitaux Universitaires de Genève
©2016 Hôpitaux Universitaires de Genève
Future of theranostics
Evolution of medical imaging
68
Fluorescence imaging
GA-Bombesin receptor antagonist
•
•
•
Ga-labeled Bombesin analog is a
synthetic bombesin receptor
antagonist, which targets gastrinreleasing peptide receptors (GRPr)
149Tb-DOTA-bombésine
161Tb-DOTA-bombésine
152Tb-DOTA-bombésine
(beta+ emitter)
©2016 Hôpitaux Universitaires de Genève
©2015 Hôpitaux Universitaires de Genève
Evolution of medical imaging
Evolution of medical imaging
Fluorescence imaging
PET & Fluorescence imaging
Synthesis of fluorescent-PET probes
Pre-op PET/MR
MRI
PET
Intra-op Fluorescent
Neuroonkologisches Zentrum
Clemenshospital GmbH
Courtesy Prof. Van Dam UMCG
(Nat. Med. 2011 17:1315-9)
©2015 Hôpitaux Universitaires de Genève
©2015 Hôpitaux Universitaires de Genève
orb light in the visible spectral region
epth penetration of light is limited to
s, thereby only allowing the treatment
al lesions. However, advances in optienabled the development of optical
corporated into endoscopes, bronchoopes to allow for the delivery of light
ities, thereby extending the scope of
DT is being explored in the treatment
luding skin,104 bladder,105 prostate,106
08
pancreatic,109 stomach,110 and head
name a few.
d in PDT can functionally be classified
active (Fig. 3). Passive PDT nanofor photosensitizers and can be made
dable material or non–polymer-based
ceramic and metallic nanoparticles.
particle carriers, made from PLGA or
wn to provide an alternative solution to
eir ability to encapsulate photosensirrier capacity. This is important as
highly hydrophobic with inherent poor
ulting in aggregation in solution that
o be parentally administered. In addigy and composition of the polymer
ized for the controlled degradation of
ce release of the photosensitizer moleer-loaded nanoparticles have been
r photoactivity than “free” photosensismaller nanoparticle carriers have a
fect compared with larger carriers due
of intracellular uptake via endocytosis,
ease of photosensitizers within the
extracellular environment. In addition,
oparticle size, the larger the surface
o, which increases the surface area
rounding medium, thus resulting in
er release rates.112 Nonbiodegradable
loaded with photosensitizers and have
nic polymeric nanoparticles, including
ontrol over size, shape, and porosity;
anges in pH and microbial attack. In
easily functionalized for selective tarue, which will allow for the selective
tosensitizers at the site of cancer while
lation of photosensitizers in nontarget
will therefore lower the concentration
used to generate the same phototoxic
sing the phototherapeutic index. Two
yes can convert low-energy radiation
missions, which can be directly transxygen to generate singlet oxygen. The
tem is that it can be activated in deep
Future of theranostics
Future of theranostics
Optical imaging
Nanotheranostics
Figure 2: WL, NIR and overlay images of catheter-based upper
GI endoscopy from OE-33 (panel A, B and C), OE-19 (panel D,
E and F) and control (panel G, H and I) groups. Upper GI
endoscopy demonstrates significant elevated NIR signal at the
tumor foci while in the control animals the signal is absent in
this region.
©2015 Hôpitaux Universitaires de Genève
©2015 Hôpitaux Universitaires de Genève
Future of theranostics
Future of theranostics
Nanotheranostics
Nanotheranostics
Nanooncology: The Future of Cancer Diagnosis and Therapy
Nanooncology: The Future of Cancer Diagnosis and Therapy
TABLE 2.
Examples of Nanoparticles Used in Cancer Imaging
IMAGING MODALITY DESCRIPTION OF NANOPARTICLE
CANCER IMAGED BY THE NANOPARTICLE
MRI
Liver tumors (ie, hepatocellular carcinoma,
liver metastases)
Superparamagnetic iron oxide nanoparticles
High-grade glioma
Ultrasmall superparamagnetic iron oxide nanoparticle Preoperative staging of pancreatic cancer
STAGE OF DEVELOPMENT/CLINICAL
TRIAL NO.
Currently used in clinical practice142
NCT00769093
NCT00920023
Pelvic lymph node metastases from prostate, NCT00147238
bladder, or other GU cancers
CT
Heavy metal (ie, gold, lanthanide, and
tantalum) nanoparticles
Solid organ tumors
Preclinical stage of development143
SPECT
TC-99m sulfur colloid nanoparticles
Sentinel lymph node mapping in invasive
breast cancer
NCT00438477
124
PET
Melanoma and malignant brain tumors
NCT01266096
Optical
Surface-enhanced Raman scattering nanoparticles
I-labeled cRGDY silica nanoparticles
Colorectal cancer
Preclinical stage of development57
Photoacoustic
Single-walled carbon nanotubes
Solid organ tumors
Preclinical stage of development144
FIGURE 2. The Criteria Nanoparticles Need to Fulfill to Be Effective Carriers for
Chemotherapeutic Drugs. (A) The nanoparticle carrier must bind or contain the desired
chemotherapeutic drug(s). (B) The nanoparticle-drug complex must remain stable in
the serum to allow for the systemic delivery of the drug. (C) The nanoparticle-drug
complex must be delivered only to tumor cells. (D) The nanoparticle must be able to
release the drug once at the site of the tumor. (E) After drug delivery, the residual
nanoparticle carrier must be safely degraded.
MRI indicates magnetic resonance imaging; NCT, National Clinical Trial; GU, genitourinary; CT, computed tomography; SPECT, single-photon emission computed
tomography; TC-99m, technetium-99m; PET, positron emission tomography; 124I, iodine-124; cRGDY, cyclic Arg-Gly-Asp-Tyr.
FIGURE 2. The Criteria Nanoparticles Need to Fulfill to Be Effective Carriers for Chemotherapeutic Drugs. (A) The nanoparticle carrier must bind or contain the desired chemotherapeutic drug(s). (B) The nanoparticle-drug complex must remain stable in the serum to allow for the systemic delivery of the
©2015 Hôpitaux
de Genève
drug. (C) The nanoparticle-drug
complexUniversitaires
must be delivered
only to tumor cells. (D) The nanoparticle must be able to release the drug once at the site
©2015 Hôpitaux Universitaires de Genève
at the target site. Examples include gadolinium-containing
monomers that assemble in cells via thiol-sensitive reduction
of 1,2-aminothiol and 2-cyanobenzothiazole and probes with
a motif sensitive to proteases such as furin and caspase-3,
which are overexpressed in tumor cells.153
Although a lot of work is currently being undertaken
preclinically to develop new nanoparticle agents, superparamagnetic iron oxide nanoparticles (SPIONs) are already
being used in clinical practice for hepatic, cardiovascular,
cellular, and lymphatic imaging. Iron oxide (magnetite,
Fe3O4; maghemite, Fe2O3) nanoparticles become superparamagnetic at room temperature if their core diameter is
20 nm or less,154 which allows for susceptibility effects at
micromolar concentrations that modify the T2 and T2*
relaxation times of water protons for enhanced MRI
contrast.155 SPIONs are also considered to have low
toxicity in vivo
as they are thought to be biodegradable,
Nanotheranostics
with the iron from the nanoparticles
released upon
CA CANCER J CLIN 2013;63:395–418
degradation into the normal plasma iron pool, where it can
subsequently be incorporated into hemoglobin in erythrocytes or used for other metabolic processes.156,157 SPIONs
have been used to characterize liver lesions since they
are phagocytosed by cells of the RES. As normal liver
parenchyma contains RES, they will accumulate SPIONs,
resulting in a decrease in signal intensity on both
T2-weighted and T1-weighted images. In contrast, most
liver tumors do not contain RES and hence they will not
uptake SPIONs, thereby improving contrast between the
tumor (high signal) and the surrounding tissue (low
signal).158 However, these signal characteristics are reversed
when SPIONs are combined with ligands for active
targeting.159 In these circumstances, SPIONs will now
of the tumor. (E) After drug delivery, the residual nanoparticle carrier must be safely degraded.
accumulate at the site of the tumor, resulting in a low
signal compared with the background liver parenchyma;
however, this relies on SPIONs avoiding the RES. To
avoid the RES and improve colloidal stability and bioAlternatively, if a nonbiodegradable material is used, it primarily composed of amphipathic phospholipids encloscompatibility, SPIONs used for active targeting are usually
must be proven to be safe at the doses needed or clear ing an interior aqueous space. They are able to contain
coated with a polymer (ie, dextran, starch, or PEG).159
hydrophilic drugs, which can remain encapsulated in the
from the subject.
Ligands such as folate are then conjugated to SPIONs via
central aqueous interior, and can be designed to adhere to
160,161
162
or PEG.
their polymer coatings of either dextran
The Nanoparticle-Drug Complex
cell membranes and release drugs after endocytosis. Studies
Folate has been used as a ligand since folate receptors are
Nanoparticles that are used as carriers will either bind the have shown improved pharmacokinetics and pharmacodyexpressed in limited quantities on the apical surfaces of
drug on their surface or entrap and encapsulate the drug to namics of drugs associated with liposomes.15 Over the
normal epithelial cells but are generally overexpressed in
protect it from degradation or denaturation. Nanoparticle years, liposomes have been surface modified with glycocarriers also offer the potential to codeliver 2 or more drugs lipids and/or polyethylene glycol (PEG) to prevent their
cancerous tissues due to the vital role that folate plays in
simultaneously for combination therapy. Newer rapid clearance from the circulation system by mononuclear
cellular proliferation. Transferrin has also been covalently
applications also include the delivery of noncytotoxic phagocytic cells from the reticuloendothelial system
coupled to SPIONs163 as it will bind to the transferrin
prodrugs that can be activated once they are delivered to (RES).16 The addition of PEG or other hydrophilic conjureceptor (also known as CD71), which is a type II
cancer cells (ie, platinum [Pt]-based chemotherapeutic gates to the surface of all types of nanoparticle carriers,
transmembrane glycoprotein that is overexpressed on the
agents can be photoreduced using visible light from their Pt including liposomes, provides increased stability of the
surfaces of proliferating cancer cells because of their
Nanotheranostics
[IV] prodrug state to the active Pt [II] anticancer
drug once nanoparticle in biological fluids while also creating a
increased iron requirements.164 SPIONs have also been
delivered inside cells using nanoparticle carriers).14 There dynamic cloud of hydrophilic and neutral chains at the surNanooncology: The Future of Cancer Diagnosis and Therapy
combined with peptide sequences such as arginylare several types of nanoparticle systems that have been face that reduces protein opsonization thereby enabling
glycyl-aspartic acid (RGD),165 which can combine with
used as carriers including liposomal, solid lipid, polymeric, nanoparticles to partially evade the macrophages of the
living
subjects.
exciting results
surface
of Nevertheless, thesemesoporous
integrins such as avb3 that are expressed oninthe
silica, and inorganic nanoparticles.
RES.2 This will increase nanoparticle half-life in blood,
provide great promise for the future.
proliferating endothelial cells such as those
undergoing
Liposomes are a biologically based nanoparticle system which combined with their ability to conjugate targeting
Carbon nanotubes (CNTs) have been studied for photofrom optia self-assembling concentric lipid bilayer that is moieties, will allow them to preferentially accumulate at
with
angiogenesis.166 Initially, SPIONs conjugated
acoustic
andmonooptical imaging since they made
have a strong
cal absorbance
in the high-near infrared region of the
clonal antibodies were not considered practical
for in vivo
electromagnetic spectrum (ie, 700-1100
398 nm), where
CA: A biologCancer Journal for Clinicians
diagnostics due to the large particle size, which
facilitated
ical systems
have a transparent window.207 This therefore
makes them
for near-infrared photothermal ablation
thisidealhas
their rapid clearance by the RES.159 However,
therapy, with the temperature within tumors shown to
proved not to be the case, with several studies showing
increase in a light-dependent and CNT dose-dependent
monoclonal antibody-conjugated SPIONs manner
having
(Fig.strong
8).196,208 In addition, CNTs are being investigated for against
their use in gene and drug delivery, since they
specificity for antigen-expressing tissues. Antibodies
can readily cross biological barriers.207 Although the mechEGFR have been conjugated with SPIONsanism
for by
thewhich
detecCNTs are internalized by cells is not fully
209
tion of colorectal, small cell lung, and esophageal
squamous
they can enter cells independently of cell
understood,
167-169
and surface
functional groups. Due to the capacity of
cell
carcinomas
in
experimental type
models.
Future of theranostics
408
CA: A Cancer Journal for Clinicians
FIGURE 3. Nanoparticles in Photodynamic Therapy. Nanoparticles can
deliver light-activatable chemicals, known as photosensitizer molecules,
to tumor cells for use in photodynamic therapy. After the absorption of
light, photosensitizer molecules can generate cytotoxic oxygen-based
reactive species, which can subsequently cause cellular damage and cell
FIGURE
3. Nanoparticles
death
via oxidative
stress. in Photodynamic Therapy. Nanoparticles can deliver
light-activatable chemicals, known as photosensitizer molecules, to tumor cells
for useby
in photodynamic
the absorption
of light,(750-1000
photosensitizer
tissues
light in thetherapy.
tissue After
transparent
window
molecules can generate cytotoxic oxygen-based reactive species, which can
nm),
which has deeper tissue depth penetration. Neverthesubsequently cause cellular damage and cell death via oxidative stress.
their backbone to form supramolecular complexes, CNTs
have been conjugated with chemotherapeutic drugs such as
doxorubicin,210 methotrexate,211 paclitaxel,212 cisplatin,213
and gemcitabine.214 Several groups have also used CNTs
for antitumor immunotherapy, whereby CNTs act as antigen-presenting carriers to improve weakly immunogenic
tumor-based peptides/antigens to trigger a humeral
215
FIGURE 8. Theranostic Nanoparticles. Nanoparticles can be designed to immune response within the patient against the tumor.
be simultaneously
used 8.
for Theranostic
diagnosis and treatment.
Using near-infrared
FIGURE
Nanoparticles.
Nanoparticles
can be
designed
Cationic
CNTs
have also been used as molecular transportlaser light, carbon nanotubes can be detected using photoacoustic imagers Using
applicable
to be
simultaneously
used for
diagnosis
and treatment.
near- for siRNA therapeutics to silence
ing in addition
to causing
tumor cell thermal
ablation
via photothermal
therapy.
geneusing
expression in both cell culture and in xenograft
infrared laser light, carbon nanotubes can be detected
216
micecell
models.
photoacoustic imag- ing in addition to causing tumor
thermal
targeting,ablation
which via
coencapsulates
Gold nanoparticles that are used for optical and
photothermalSPIONs
therapy. for imaging
and doxorubicin for controlled drug release.204 Other photoacoustic imaging can also be used in PTT. Following
platforms include using SPION cores with a polycationic irradiation, the high electron density within the metallic
surface coating (ie, poly(hexamethylene biguanide) or lattice of gold nanoparticles results in absorption of photon
polyethyleneimine), which can bind siRNA through energy that, in turn, causes the lattice and hence the
electrostatic interactions to form magnetic vectors that can nanoparticle to heat up. The small size and the rapid
be rapidly drawn to and concentrated on the surface of the heating ability of gold nanoparticles are attractive for the
target cells using the attractive force of an externally selective heating and killing of cancer cells with an
applied magnetic field. This facilitates the uptake of the appropriate light source without the destruction of the
magnetic vector into the cell endosomes, thereby improving surrounding normal and healthy tissue. Although NIRsiRNA transfection efficiency.205 SPIONs have also been mediated ablation has shown promise, its efficacy is
radiolabeled with 64Cu (for combined imaging PET/MRI), limited by its depth of penetration, which only allows the
conjugated with doxorubicin (for chemotherapy), and treatment of superficial tumors up to 2 to 3 cm. However,
functionalized with RGD (for tumor vasculature RF ablation may be able to overcome this obstacle and
targeting).206 As PET has excellent sensitivity but relatively allow the treatment of deep-seated tumors since gold
poor spatial resolution, its combination with MRI will nanoparticles have been shown to interact with shortwave
provide excellent spatial resolution and soft-tissue contrast RF waves to produce heat.217 Currently, RF treatments use
that is superior to CT while also not delivering any of macroscopic electrodes to induce ablation, which is painful
ionizing radiation associated with CT to the patient. and can cause damage to surrounding tissues. However, the
Furthermore, the conjugation of doxorubicin onto use of microelectrodes could make this technique less
PEGylated SPIONS was performed via pH-sensitive invasive and more effective provided that nanoparticles can
hydrazone bonds, thereby allowing controlled drug release be concentrated above a threshold level at the site of the
within the acidic microenvironment of tumors. Although tumor.100 Multimodal nanoparticles have also been created
these and many other elegant nanoplatform designs have such as those that have a superparamagnetic core to allow
been tested within cell culture, they have yet to be validated imaging with MRI, and a gold shell to allow PTT.218
less, the dye’s toxicity remains a major problem. Entrapping
the dye in a nanoparticle carrier, which is biologically inert,
can therefore reduce its toxicity to normal tissue while
allowing PDT penetration in deeper tissues. Other groups
are also exploring the use of exciting photosensitizers
(energy acceptors) indirectly through fluorescence resonance energy transfer from photon absorbing dyes (energy
donors).113 By physically encapsulating the dye and the
photosensitizer in the same nanoparticle, this approach
allows for the efficient transfer of energy between the
dye, which acts Molecular
as an intermediary,
and the active
imaging
coencapsulated photosensitizer. For efficient photon excitation using this concept, the loading density of the energydonating photon absorption dye needs to be much higher
than that of the energy-accepting photosensitizer. Hence,
• silica
The
role of
therapy
inbionuclear medicine was one of its
modified
nanoparticles
have been
used as they are
compatible, stable without releasing encapsulated hydroearliest applications
phobic molecules, and suitable for PDT because their
114
412
CA: A Cancer Journal for Clinicians
porous matrix is permeable to oxygen molecules.
• As
diagnostic
increased exponentially,
Active
PDT our
nanoparticles
can themselvescapability
generate
reactive species
the presence
of a photosensitizer.
we without
moved
from
an emphasis on diagnosis to an
This was first appreciated by Samia et al, who found that in
emphasis
on therapy
addition to
sensitizing photosensitizer
molecules through a
fluorescence resonance energy transfer, semiconductor QDs
We generate
are now
approaching
the ideal combination of
could•themselves
singlet oxygen
alone via a triplet
energy transfer
without the need forcapability
photosensitizers, in both the diagnostic and
extraordinary
albeit with a lower efficiency.115 Other groups have also
therapeutic
aspects
ofanthe field
investigated
the ability of nanoparticles
to play
additional active intermediary role in the process of PDT,
in addition
to encapsulating photosensitizers
and has
targetingbecome a vital part of
• Molecular
imaging
them to cancer cells.101 These nanoparticles will emit
personalized medicine
luminance of an appropriate wavelength to active
©2015 Hôpitaux Universitaires de Genève
Conclusions
VOLUME 63 _ NUMBER 6 _ NOVEMBER/DECEMBER 2013
©2016 Hôpitaux Universitaires de Genève
Future of theranostics
Gold nanoparticles (GNPs) are readily synthesized structures that
absorb light strongly to generate thermal energy which induces
photothermal destruction of malignant tissue.
©2015 Hôpitaux Universitaires de Genève
Conclusions
Theranostics
• The field of Theranostics is facilitating the shift from
'trial and error' medicine to personalized medicine
• Theranostics helps identifying and selecting patients
with a particular molecular phenotype indicative of
positive response to treatment
• The focus of theranostics lies on imaging biomarkers
that can identify patients who will benefit from
molecularly targeted therapy and are going to fail to
respond to standard treatment.
403
©2016 Hôpitaux Universitaires de Genève
Conclusions
Shift of paradigm
« Moving from standard treatment
of a disease to specific treatment
of individual patients »
©2016 Hôpitaux Universitaires de Genève
Téléchargement