Vous êtes sur la page 1sur 22

US 20030028071A1

(19) United States


(12) Patent Application Publication (10) Pub. No.: US 2003/0028071 A1
Handy et al. (43) Pub. Date: Feb. 6, 2003

(54) THERMOTHERAPY VIA TARGETED (22) Filed: Jun. 18, 2002


DELIVERY OF NANOSCALE MAGNETIC
PARTICLES Related US. Application Data

(75) Inventors: Erik Schroeder Handy, Arlington, MA (60) Provisional application No. 60/307,785, ?led on Jul.
(US); Robert Ivkov, Marblehead, MA 25, 2001.
(US); Diane Ellis-Busby, Lancaster,
MA (US); Allan Foreman, Epping, NH Publication Classi?cation
(US) (51) Int. Cl.7 ..................................................... .. A61N 2/00
Correspondence Address: (52) US. Cl. .............................................................. .. 600/12
ALTERA LAW GROUP, LLC (57) ABSTRACT
6500 CITY WEST PARKWAY
SUITE 100 Disclosed are compositions comprising magnetic nanopar
MINNEAPOLIS, MN 55344-7704 (US) ticles, a biocompatible coating, and a target-speci?c ligand.
Also disclosed are devices for treating diseased tissue for
(73) Assignee: Triton Biosystems, Inc., Chelmsford, use With such compositions. Further disclosed are methods
MA for treating diseased tissue, such as cancer, using such
compositions and devices, as Well as methods for treating
(21) Appl. No.: 10/176,950 diseased tissue utilizing hypertermia.

101
102

104 /105
I I /\ x 106
120 — I l— /

uI zu 0
Patent Application Publication Feb. 6, 2003 Sheet 1 0f 7 US 2003/0028071 A1

H LWIH IW HT HI gwa/
9
\ 106

Figure 1.
Patent Application Publication Feb. 6, 2003 Sheet 2 0f 7 US 2003/0028071 A1

mmn
Execs
won

N8www86 E.mzmE

@5 2 {25oz wwm

m5

b29056 3:950 / 0mm

m5
Patent Application Publication Feb. 6, 2003 Sheet 3 0f 7 US 2003/0028071 A1

500

400

300

200

100

(A)Qir ert O

—100

-200

-300

-400

-500
0 0.5 1 1.5 2 2.5

'Fme (uSec.)

Figure 4a.

2000

1500

1000

/ \ , ,
(QmeArt) O

i \ / W
4000 \ /
4500 \/
-2000

Time (uSec.)

Figure 4b.
Patent Application Publication Feb. 6, 2003 Sheet 4 0f 7 US 2003/0028071 A1

500

400 _

3o

200

100

(CurAen)t

400

0 5000 1 0000 15000 20000


Tlmu (u See.)

Figure 5a.

1 500

1000 r ‘

500

(FMaGigenulsd)c D

-500

-1000 r

‘1500
0 5000 10000
‘l’lma (u See.)

Figure 5b.
Patent Application Publication Feb. 6, 2003 Sheet 5 0f 7 US 2003/0028071 A1

640
642

644

690

Figure 6.
Patent Application Publication Feb. 6, 2003 Sheet 6 0f 7 US 2003/0028071 A1

71s 776 f 774

768
/ 764

776 11a _/ 780

768
,764

Figure 7.
Patent Application Publication Feb. 6, 2003 Sheet 7 0f 7 US 2003/0028071 A1

840
1542

\ 844 890
/

850
848

Figure 8.
US 2003/0028071 A1 Feb. 6, 2003

THERMOTHERAPY VIA TARGETED DELIVERY [0007] Immunotherapy is a rapidly expanding type of


OF NANOSCALE MAGNETIC PARTICLES therapy used for treating a variety of human diseases includ
ing cancer. The FDA has approved a number of antibody
CROSS REFERENCE TO RELATED based cancer therapeutics. The emergence of antibody thera
APPLICATION pies is made possible by important advances in antibody
[0001] This is a non-provisional application claiming the technologies. The ability to engineer antibodies, fragments,
bene?t of and priority to provisional patent application No. and peptides With altered properties such as antigen binding
60/307,785 ?led on Jul. 25, 2001. af?nity, molecular architecture, speci?city, and valence has
enhanced their use in therapies. The advantages of antibody
TECHNICAL FIELD engineering have overcome the limitations of mouse mono
clonal antibodies. Cancer immunotherapeutics have made
[0002] The present invention relates generally to thermo use of advances in the chimeriZation and humaniZation of
therapy, more speci?cally, to magnetic material composi mouse antibodies to reduce immunogenic responses in
tions, devices for use With magnetic material compositions, humans. High af?nity human antibodies have also been
and methods related thereto for thermotherapy via targeted obtained from transgenic mice that contain many human
delivery of nanoscale magnetic particles. immunoglobulin genes. In addition, phage display technol
ogy, ribosome display, and DNA shuffling have alloWed for
BACKGROUND the discovery of antibody fragments and peptides that have
[0003] Despite considerable research effort and some suc the desirable properties of high af?nity and loW immuno
cess, cancer is still the second leading cause of death in the genicity for use as targeting ligands. All of these advances
United States, claiming more than 500,000 lives each year have made it possible to design an immunotherapy that has
according to American Cancer Society estimates. Traditional a desired antigen binding af?nity, speci?city, and minimal
treatments are either invasive or expose the patient to immune response.
considerable toxicity With often only modest success. Early [0008] The ?eld of cancer immunotherapy makes use of
detection, a result of better diagnostic practices and tech markers that are expressed or over-expressed on cancer cells
nology has improved the prognosis for many patients. Nev in comparison to normal cells. The identi?cation of such
ertheless, some cancers defy currently available treatment markers is ongoing and the choice of a ligand/marker
options, despite these improvements. Of the many forms of combination is critical to the success of any immunotherapy.
cancer that still pose a medical challenge, prostate, breast, Immunotherapy has fallen into several classes: (1) antibod
lung, and liver claim the vast majority of lives each year. ies themselves that target groWth receptors, disrupt cytokine
[0004] Breast cancer is the most common cancer in pathWays, or induce complement or antibody-dependent
Women, With Well over 100,000 neW cases being diagnosed cytotoxicity; (2) direct arming of an antibody With a toxin,
each year in the United States alone. Breast cancer usually a radionucleotide, or a cytokine; (3) indirect arming of an
begins in the cells lining a breast duct, the eventual invasion antibody by attachment to immunoliposomes used to deliver
of these cells outside the ductal environment presenting the a toxin or by attachment to an immunological cell effector
risk of metastases. Later stages of breast cancer are fatal and (bispeci?c antibodies). Although armed antibodies have
typically have advanced to a point in Which cancer cells have shoWn more potent tumor activity in clinical trials, there
invaded the lymphatic system, and may include other organs have been unacceptably high levels of toxicity. The disad
such as the liver, bone, lungs, and brain. vantage of therapies that rely on delivery of immunotoxins
or radionucleotides (direct and indirect arming) has been
[0005] Conventional treatments for breast cancer, both in that these agents are active at all times. There have been
late and early stages, typically include surgery folloWed by problems With damage to non-tumor cells and toxicity issues
radiation and/or chemotherapy. These techniques are not along With delivery challenges. Many immunotherapies
alWays effective, and even if effective, they suffer from have faced challenges With shed markers and delivery to the
certain de?ciencies. Surgical procedures include complete intended target. Cancer cells commonly shed antigen targets
removal of the breast and removal of only the tumor into the blood stream. Many antibody-based therapies are
(lumpectomy). In early stage cancer, complete removal of diluted by interaction With shed antigens. In addition,
the breast provides the best assurance against recurrence, but immune complexes can be formed betWeen the immuno
is dis?guring and requires the patient to make a very dif?cult therapeutic and the shed antigen, Which can lead to dose
choice. Lumpectomy is less dis?guring, but is associated limiting toxicities.
With a greater risk of recurrence. Radiation and chemo
therapy are arduous and are not completely effective against [0009] Generation of heat in a range of about 40° C. to
recurrence. about 46° C. (hyperthermia) can cause irreversible damage
to diseased cells, Whereas normal cells are not similarly
[0006] For these reasons, it Was desirable to provide affected. Diseased tissue may be treated by elevating the
improved and alternative techniques for treating breast can temperature of the individual cells contained Within to a
cer. Such techniques should be less invasive and traumatic
lethal level (cellular thermotherapy) using a suitable mag
to the patient than the present techniques, should result in netic material con?ned to the vicinity of the cell and
minimum or no dis?gurement of the breast, and should be
induction heating the material using an alternating magnetic
effective locally Within target sites of the breast and other ?eld
diseased organs. Preferably, the techniques should be
capable of being performed in a single or very feW treatment [0010] Hyperthermia may hold promise as a treatment for
session(s), With minimal toxicity to the patient. In addition, cancer because it induces instantaneous necrosis (typically
the diseased tissues should be targeted by the treatment called thermo-ablation) and/or a heat-shock response in cells
Without requiring signi?cant operator skill and input. (classical hyperthermia), leading to cell death via a series of
US 2003/0028071 A1 Feb. 6, 2003

biochemical changes Within the cell. State-of-the-art sys bound to the particle and partially covered by the coating, or
tems that employ radio-frequency (RF) hyperthermia, such intercalated into the coating. The present invention also
as annular phased array systems (APAS), attempt to tune pertains to devices for treating cancer by interacting With
E-?eld energy for regional heating of deep-seated tumors. magnetic particles targeted to cancer cells in a patient. Such
Such techniques are limited by the heterogeneities of tissue a device includes a magnetic generator having a core de?n
electrical conductivities and that of highly perfused tissues, ing at least part of a magnetic circuit, tWo poles of the core
leading to the unsolved problems of ‘hot spot’ phenomena in de?ning a gap therebetWeen, a magnetic ?eld passing
unintended tissues With concomitant underdosage in the betWeen tWo poles, the gap being of suf?cient siZe to receive
desired areas. These factors make selective heating of spe a portion of the patient containing the cancer cells; and a
ci?c regions With such E-?eld dominant systems very dif poWer supply coupled to provide energy to the magnetic
?cult. generator so that the magnetic ?eld passing betWeen the tWo
poles alternates at a frequency of about 1 kHZ or more.
[0011] Another strategy that utiliZes RF hyperthermia
requires surgical implantation of microWave- or RF-anten [0019] The present invention further pertains to methods
nae or self-regulating thermal seeds. In addition to its related to such magnetic material compositions and devices
invasiveness, this approach provides only limited (if any) for the treatment of cancer. One such method includes the
treatment options for metastases because it requires knoWl administration to the patient of a magnetic material compo
edge of the precise location of the tumor, and is thus sition that includes at least one, single domain, magnetic
incapable of targeting undetected individual cancer cells or particle attached to a cancer-cell speci?c ligand, and appli
cell clusters not immediately adjacent to the primary tumor cation of an alternating magnetic ?eld to a region of the
site. Clinical outcomes of these techniques are limited by patient containing the cancer so as to inductively heat the
problems With deposition of physical poWer to the desired magnetic material composition and kill the cancer cells.
tumor tissues.
[0020] The therapeutic methods of the present invention
[0012] Hyperthermia for cancer treatment using colloidal provide for the treatment of diseased tissue, such as cancer,
single domain magnetic suspensions (i.e., magnetic ?uids) in a safe and effective manner, With minimal invasion, and
eXposed to RF ?elds has been recogniZed for several short treatment periods.
decades. HoWever, a major problem With magnetic ?uid
hyperthermia has been the inability to selectively deliver a [0021] The above summary of the present invention is not
lethal dose of particles to the tumor cells. intended to describe each illustrated embodiment or every
implementation of the present invention. The ?gures and the
SUMMARY OF THE INVENTION detailed description Which folloW more particularly eXem
plify these embodiments.
[0013] In vieW of the above, there is a need for a hyper
thermia based treatment method for diseased tissue, such as BRIEF DESCRIPTION OF THE DRAWINGS
cancer, that incorporates selective delivery of magnetic
?elds to the diseased tissue. It is also desirable to have [0022] The invention may be more completely understood
methods for treating diseased tissue, such as cancer, in a safe in consideration of the folloWing detailed description of
and effective manner, With minimal invasion, and short various embodiments of the invention in connection With the
treatment periods. accompanying draWings, in Which:
[0014] It is, therefore, an object of the present invention to [0023] FIG. 1 schematically illustrates a thermotherapy
formulate magnetic material compositions including nanos treatment system according to an embodiment of the present
cale magnetic particles and target-speci?c ligands useful in invention;
the treatment of diseased tissue, such as cancer.
[0024] FIG. 2 schematically illustrates a thermotherapy
[0015] It is another object of the present invention to treatment according to an embodiment of the present inven
provide a device for the treatment of diseased tissue that is tion;
capable of providing an alternating magnetic ?eld.
[0025] FIG. 3 schematically illustrates a circuit for pro
[0016] It is yet another object of the present invention to ducing a thermotherapeutic alternating magnetic ?eld
provide a method that utiliZes compositions of nanoscale according to an embodiment of the present invention;
magnetic materials and target-speci?c ligands in conjunc
tion With a device that provides alternating magnetic ?elds [0026] FIG. 4a graphically illustrates a thermotherapeutic
to treat diseased tissue by killing diseased cells via hyper sinusoidal current Waveform according to an embodiment of
thermia. the present invention;
[0017] It is a further object of the present invention to [0027] FIG. 4b graphically illustrates a thermotherapeutic
provide methods for the treatment of diseased tissue, such as triangular current Waveform according to an embodiment of
cancer, in a safe and effective manner, With minimal inva the present invention;
sion, and short treatment periods.
[0028] FIG. 5a graphically illustrates a thermotherapeutic
[0018] The present invention pertains to magnetic material sinusoidal Waveform modulation according to an embodi
compositions that include magnetic particles forming a ment of the present invention;
single magnetic domain; a biocompatible coating material
for the particle; and a ligand that is selective to at least one [0029] FIG. 5b graphically illustrates a thermotherapeutic
cancer marker, and that can be bound to an uncoated portion pulsed Waveform modulation according to an embodiment
of the particle, bound to a coated portion of the particle, of the present invention;
US 2003/0028071 A1 Feb. 6, 2003

[0030] FIG. 6 schematically illustrates a thermotherapeu 108. The AMF generator produces an AMF in the magnetic
tic bioprobe con?guration according to an embodiment of circuit 102 that exits the magnetic circuit at one pole face
the present invention; 104, passing through the air gap and the desired treatment
area of the patient, and reenters the circuit through the
[0031] FIG. 7 schematically illustrates a disease speci?c
opposing pole face 104, thus completing the circuit. An
targeting ligand component of a bioprobe according to an operator or medical technician is able to both control and
embodiment of the present invention; and monitor the AME characteristics and bed positioning via the
[0032] FIG. 8 schematically illustrates disease speci?c control panel 120.
bioprobes bound to a disease cell surface according to an
[0036] FIG. 2 illustrates a treatment of a patient With a
embodiment of the present invention.
device for treating cancer according to an embodiment of the
[0033] While the invention is amenable to various modi present invention. The diseased or cancerous treatment area
?cations and alternative forms, speci?cs thereof have been 205 of the patient is localiZed in the region betWeen the
shoWn by Way of example in the draWings and Will be magnetic poles 204 via the positionable bed 206. This region
described in detail. It should be understood, hoWever, that may be the breast, an area in close proximity to the breast,
the intention is not to limit the invention to the particular lymph nodes and ducts, liver, lung, bones, or any other site
embodiments described. On the contrary, the intention is to or region of metastasis. An AMF may be applied to the
cover all modi?cations, equivalents, and alternatives falling treatment area 205 of the patient, as illustrated by the
Within the spirit and scope of the invention as de?ned by the magnetic lines of ?ux 212. The magnetic ?eld, manifested
appended claims. by the magnetic lines of ?ux 212 interacts With both healthy
and diseased tissue in the localiZed area. Bioprobes 210,
DETAILED DESCRIPTION containing at least one appropriate ligand selective to the
particular type of cancer, are bound to cancer cells 214. In
[0034] The present invention pertains to magnetic material the illustrated case, the bioprobes 210 are selective to breast
compositions, devices for treating diseased tissue for use cancer. The bioprobes 210 become excited by the interacting
With magnetic material compositions, and methods for treat applied AMP and are inductively heated to a temperature
ing diseased tissue utiliZing such devices and magnetic suf?cient to kill diseased or cancerous cells. Heat generated
material compositions. The therapeutic methods disclosed in the excited the bioprobes 210 passes to the cancer cells,
herein include the targeted delivery of nanometer siZed thereby causing the cancer cells to die.
magnetic particles to diseased tissue. Speci?cally, the
present invention pertains to such compositions, devices, [0037] It Will be appreciated that other types of cancers
and methods for the treatment of cancer. The term “bio may be treated using the device, for example, lung, prostate
probe”, as used herein, refers to the composition including and the like. Furthermore, the poles 204 may be formed from
a magnetic particle, a biocompatible coating material, and a pieces Whose gap is adjustable, so as to permit other parts of
target-speci?c ligand. The term “ligand”, as used herein, the body to be treated. It is advantageous to set the gap
refers to compounds Which target biological markers. betWeen the poles 204 to be suf?ciently large to permit the
Examples of ligands include proteins, peptides, antibodies, part of the body containing the cancer to enter the gap, but
antibody fragments, saccharides, carbohydrates, glycans, not be so large as to reduce the magnetic ?eld strength.
cytokines, chemokines, nucleotides, lectins, lipids, recep [0038] FIG. 3 illustrates a circuit for producing an AMF
tors, steroids, neurotransmitters, Cluster Designation/Differ according to an embodiment of the present invention. The
entiation (CD) markers, imprinted polymers, and the like. AMF generator 318 is supplied With alternating current
The methods for treating diseased tissue disclosed herein
include administering to a patient the bioprobes suspended (AC) poWer via the conduit 316. A circulating ?uid supply
is also provided in the conduit 316. The AMF generator 318
in an appropriate medium, and applying, via a device
may become hot, and it may be cooled With the circulating
capable of interacting With the bioprobes, an alternating
?uid supply While in operation. The ?uid may be Water;
magnetic ?eld to an area of the patient containing the
hoWever a ?uid such as silicone oil or other inorganic or
bioprobes to heat the bioprobes sufficiently to kill cancerous
cells. organic (carbon based) ?uids With suitable thermal and
electric properties may be preferable. Such a ?uid increases
[0035] One embodiment of the invention, as illustrated in generator ef?ciency. The energy produced by the generator
FIG. 1, includes an alternating magnetic ?eld gen 318 is directed through the AME matching netWork 320
erator located Within a cabinet 101 designed to produce an Where the impedance of the generator is matched to the
alternating magnetic ?eld that may be guided to a impedance of the coil 322. The impedance of the AME
speci?c location Within a patient 105 by a magnetic circuit matching netWork 320 may be adjustable to minimiZe the
102. The therapeutic methods of the present invention may energy re?ected back to the generator 318. In another
be performed folloWing a diagnosis or evaluation of cancer embodiment, the generator frequency may be automatically
or a pre-cancerous condition or other disease in one or more adjusted to minimiZe the re?ected energy. The modi?ed
areas of the patient. Speci?cally, the diagnosed disease may energy may be directed to the magnetic circuit 302. An AMF
be breast cancer or metastatic breast cancer. The manner of is induced in the magnetic circuit 302 as a result of the
making the diagnosis does not form part of the invention and current passing through the solenoid coil 322. Magnetic
may be performed using any standard method. HoWever, the lines of ?ux 312 are produced in the gap 333 betWeen the
present invention, or aspects thereof, may be amenable to a poles 304 in the magnetic circuit 302. A feedback loop 324
diagnostic function alone or in conjunction With another may be provided for monitoring the magnetic ?eld pro?le in
method or apparatus. The patient lies upon an X-Y horiZon the gap 333 betWeen the poles 304. A suitable probe 354
tal and vertical axis positioning bed 106. The bed 106 is both provides data to the monitor 352, Which relays information
horiZontally and vertically positionable via a bed controller to the controller 356 via an appropriate data bus 324.
US 2003/0028071 A1 Feb. 6, 2003

Information from the controller 356 is relayed to the gen [0044] The AMF may be constrained to prevent heating
erator 318 via an appropriate data bus 358. Monitoring the healthy tissue to lethal temperatures (typically§43° C.).
magnetic ?eld pro?le may be useful in detecting the pres This may be accomplished in a variety of Ways.
ence of magnetic particles, monitoring an inductance of
tissue, and monitoring the temperature of tissue located in [0045] The peak amplitude of the AMP may be
the gap 333. adjusted.
[0039] Measuring alternating magnetic ?elds directly is [0046] The PRF may be adjusted.
extremely dif?cult. Because the AMP is proportional to the
current in the coil 322, characteristics of the AMP may be
[0047] The pulse Width may be adjusted.
de?ned in terms of the coil current, Which can readily be [0048] The fundamental frequency may be adjusted.
measured With available test equipment. For eXample, the
coil current may be vieWed and measured With a calibrated [0049] These four characteristics may be adjusted to maXi
RogoWski coil and any oscilloscope of suitable bandWidth. miZe heating rate of the bioprobes and, simultaneously, to
The fundamental Waveform may be observed as the direct minimiZe the heating rate of the healthy tissue located Within
measure of the magnitude and direction of the coil current. the treatment volume. These conditions may vary depending
Many different types of fundamental Waveforms may be upon tissue types to be treated, thus the operator may
used for the AMP. For eXample, FIG. 4a illustrates a determine ef?cacious operation levels. In one embodiment,
sinusoidal current Waveform, and FIG. 4b illustrates a one or more of these characteristics may be adjusted during
triangular current Waveform. The shape of the fundamental treatment based upon one or more continuously monitored
Waveform may also be square, saWtooth, or trapeZoidal. physical characteristics of tissue in the treatment volume by
the probe 354, such as temperature or impedance. This
[0040] Most practical generators produce an approxima information may then be supplied as input to the generator
tion of these Waveforms With some amount of distortion. In
318, via the monitor 352, the data bus 324, the controller
most applications, this Waveform may be nearly symmetri 356, and the data bus 358 to control output, constituting the
cal around Zero, as illustrated in FIGS. 4a and 4b. HoWever,
feedback loop. The generator output may be adjusted so that
there may be a static (or DC) current superimposed on the the peak AMF strength is betWeen about 10 and about
Waveform (DC offset). FIGS. 4a and 4b shoW at least one
cycle of tWo different fundamental Waveforms With Zero or
10,000 Oersteds (Oe). Preferably, the peak AMF strength is
betWeen about 20 and about 3000 Oe, and more preferably,
near Zero DC offsets. The fundamental period may be
betWeen about 100 and about 2000 Oe.
de?ned as the time it takes to complete one cycle. The
fundamental frequency may be de?ned as the reciprocal of [0050] In another embodiment of the present invention,
the fundamental period. The fundamental frequency may be the differential heating of the bioprobes, as compared to that
betWeen 1 kHZ and 1 GHZ, preferably betWeen 50 kHZ and of the healthy tissue, may be maXimiZed. The bioprobes 210
15 MHZ, and more preferably betWeen 100 kHZ and 500 heat in response to each cycle of the AMP. Assuming the
kHZ. The fundamental frequency may be intentionally fundamental frequency, the PRF, and the pulse Width remain
modulated (as in many high-resolution RADAR designs), constant, the heat output of the bioprobe 210 Will continue
and may often vary slightly as a result of imperfections in to increase as peak amplitude of the AMP increases until the
the RF generator design. magnetic material of the bioprobe reaches saturation.
[0041] The amplitude of the Waveform may also be modu Beyond this point, additional increases in AMF amplitude
lated. FIG. 5a illustrates an embodiment in Which a sinu yield almost no additional heating. At AMF amplitudes
soidal current modulation envelope may be used, and FIG. beloW saturation hoWever, it can be said that bioprobe
5b illustrates an embodiment that utiliZes a square modula heating is a function of AMP amplitude. Unlike bioprobes,
tion envelope. The shape of the amplitude modulation healthy tissue heating is a result of eddy current How and a
envelope may typically be sinusoidal, square, triangular, function of the rate of change of the AMP. In particular, the
trapeZoidal or saWtooth, and may be any variation or com eddy current and resultant tissue heating folloWing the
bination thereof, or may be some other shape. expressions:
[0042] The AMF produced by the generator may also be [0051] (1) reddy dB/dT
pulsed. Pulse Width is traditionally de?ned as the time
betWeen the —3 dBc points of the output of a square laW [0052] (2) Tissue Heating Ieddy2
crystal detector. Because this measurement technique is [0053] From the relationships (1) and (2), it is evident that
cumbersome in this application, We use an alternate de?ni reducing the rate of change of the AMP yields a signi?cant
tion of pulse Width. For the purpose of this invention, pulse reduction in tissue heating. In one embodiment of the
Width may be de?ned as the time interval betWeen the 50% present invention, this relationship is eXploited by using a
amplitude point of the pulse envelope leading edge and the symmetrical triangular Wave, as shoWn in FIG. 4b, as the
50% amplitude point of the pulse envelope trailing edge. fundamental Waveform. By avoiding the high rates of
The pulse Width may also be modulated. change as a sinusoid crosses the X-aXis (FIG. 4a), and
[0043] The pulse repetition frequency (PRF) is de?ned as substituting the constant but loWer rate of change associated
the number of times per second that the amplitude modu With a triangular Waveform (FIG. 4b), tissue heating may be
lation envelope is repeated. The PRF typically lies betWeen reduced With little or no sacri?ce in bioprobe heating. A
0.0017 HZ and 1000 MHZ. The PRF may also be modulated. triangular Waveform, as shoWn in FIG. 4b, may be achieved
The duty cycle may be de?ned as the product of the pulse by using an appropriate generator, such as a linear ampli?er
Width and the PRF, and thus is dimensionless. In order to be based generator. Some distortion of the triangle is inevitable,
de?ned as pulsed, the duty of the generator 318 must be less but tangible reductions in tissue heating result from even
than unity (or 100%). small reductions in dB/dT.
US 2003/0028071 A1 Feb. 6, 2003

[0054] The heating of both the tissue and the bioprobes may be shaped to in?uence the ?ux path through gap 333.
increase With increased AMF amplitude. At loW AMF ampli The pole faces 304 may be detachable and may be chosen to
tudes, small increases yield signi?cant increases in magnetic extend the magnetic circuit 302 as much as possible, to
heating. As the bioprobes approach saturation however, their minimiZe gap the 333 While leaving suf?cient space to
relationship With the AMP amplitude becomes one of dimin receive that portion of the patient being treated.
ishing return. This relationship is unique to the particular
[0057] FIG. 6 discloses a bioprobe con?guration accord
magnetic material, as are the values that constitute “loW” or
ing to an embodiment of the present invention. A spherical
“saturating” AMF amplitudes. Bioprobe heating is at ?rst shaped bioprobe 690, having a magnetic particle 642 at its
related to the AMP amplitude by an exponent >1, Which
center, is shoWn. The magnetic particle 642 may be covered
gradually diminishes to an exponent <1 as saturation is
With a coating 644. At least one targeting ligand 640, such
approached. At typical pulse Widths and duty cycles, eddy as, but not limited to, an antibody, may be located on an
current heating is directly related to duty cycle. The capa
bility to pulse the generator output, as illustrated in FIGS. 5a
exterior portion of the bioprobe 690. The targeting ligand
640 may be selected to seek out and bond With a particular
or 5b, alloWs the bene?ts of operating at higher AMF
type of diseased or cancerous cell. Heat is generated When
amplitudes While maintaining a constant reduced tissue
the magnetic particle 642 of the bioprobe 690 is subjected to
heating by reducing the duty cycle. the AMP. In a general sense, this heat represents an energy
[0055] It is desirable to apply the AMP to the treatment loss as the magnetic properties of the material are forced to
area 205 of the patient 105. Generating high peak amplitude oscillate in response to the applied alternating magnetic
AMF over a large area requires a very large AMF generator ?eld. The amount of heat generated per cycle of magnetic
and exposes large amounts of healthy tissue to unnecessary ?eld and the mechanism responsible for the energy loss
eddy current heating. Without some means of directing the depend on the speci?c characteristics of both the magnetic
?eld to Where it is useful, disease in the chest or trunk could material of the particle 642 and the magnetic ?eld. The
only be practically treated by placing the patient Within a magnetic particle 642 heats to a unique temperature, knoWn
large solenoid coil. This Would expose most of the major as the Curie temperature, When subjected to the AMP. The
organs to eddy current heating, Which must then be moni Curie temperature is the temperature of the reversible fer
tored and the AMP adjusted so as not to overheat any part romagnetic to paramagnetic transition of the magnetic mate
of a variety of tissue types. Each of these tissue types has a rial. BeloW this temperature, the magnetic material heats in
different rate of eddy current heating. The peak AMF an applied AMF. HoWever, above the Curie temperature, the
strength Would need to be reduced to protect those tissue magnetic material becomes paramagnetic and its magnetic
types that experience the most extreme eddy current heating. domains become unresponsive to the AMP. Thus, the mate
If the varieties of exposed tissue are minimiZed, it is likely rial does not generate heat When exposed to the AMP above
that the AMP strength can be increased, and thereby reduc the Curie temperature. As the material cools to a temperature
ing the treatment time and increasing the ef?cacy. One beloW the Curie temperature, it recovers its magnetic prop
method of con?ning the high peak amplitude AMF to erties and resumes heating, as long as the AMP remains
treatment area 205 is by de?ning the loWest reluctance path present. This cycle may be repeated continuously during
of magnetic ?ux With high permeability magnetic material. exposure to the AMP. Thus, magnetic materials are able to
This path is referred to as a magnetic circuit (102 in FIG. 1 self-regulate the temperature of heating. The temperature to
and 302 in FIG. 3). The magnetic circuit may be provided Which the particle 642 heats is dependent upon the magnetic
so that all or most of the magnetic ?ux produced by the coil properties of the material, characteristics of the magnetic
322 may be directed to the treatment area 205. One bene?t ?eld, and the cooling capacity of the diseased tissue (tumor)
of the magnetic circuit 302 is that the necessary amount of site 214. Selection of the magnetic material and AMF
?ux may be reduced since the amount of ?ux extending characteristics may be tailored to optimiZe treatment ef?cacy
beyond the treatment area 205 is minimiZed. Reducing the of a particular tissue type. In an embodiment of the present
required ?ux reduces the required siZe and poWer of the invention, the magnetic material may be selected to possess
AMP generator, and minimiZes exposure of tissue outside a Curie temperature betWeen 40° C. and 150° C.
the treatment area 205 to high peak amplitude AMF. In
addition, a reduced area of AMP exposure avoids the unin [0058] The magnetic attributes of ferromagnets, ferrites
tentional heating of surgical or dental implants and reduces (ferrimagnets), and superparamagnets are determined by an
the likelihood that they Will need to be removed prior to ensemble of interacting magnetic moments in a crystalline
treatment, thereby avoiding invasive medical procedures. structure. The magnetic moments of ferromagnets are par
Concentrating the ?eld permits the treatment of large vol allel and equal in magnitude, giving the material a net
umes Within the chest or trunk With a portable siZe device. magnetiZation, or net magnetiZation vector. By contrast,
ferrites are ferrimagnetic, Where adjacent magnetic moments
[0056] The material used to fabricate the magnetic circuit are parallel in direction and unequal in magnitude, yielding
302 may be appropriate to the peak amplitude and frequency a net magnetiZation in ferrimagnetic coupling. Superpara
of the AMP. The material may be, but is not limited to, iron, magnets possess clusters or collections of atomic magnetic
poWdered iron, assorted magnetic alloys in solid or lami moments that are either ferromagnetic or ferrimagnetic,
nated con?gurations and ferrites. The pole faces 104, 204, hoWever there may be no particular relationship in the
and 304 may be shaped and siZed to further concentrate the
orientation of the moments among several clusters. Thus, a
?ux produced in the treatment area. The pole faces 304 may
superparamagnetic material may possess a net magnetic
be detachable. Different pole pieces having different siZes moment.
and shapes may be used, so that the treatment area and
volume may be adjusted. When passing from one material to [0059] A magnetic domain may be de?ned as an area of
another, the lines of magnetic ?ux 312 travel in a direction locally saturated magnetiZation, and the magnetic domain
normal to the plane of the interface plane. Thus, the face 304 boundary thickness, or the distance separating adjacent
US 2003/0028071 A1 Feb. 6, 2003

magnetic domains, may be about 100 nm. Thus, magnetic cobalt, are added to the magnetite, this siZe range can be
particles (ferromagnetic or ferrimagnetic) possessing a smaller. This results directly from the fact that cobalt gen
dimension smaller than 250 nm, and preferably less than erally possesses a larger magnetic moment than magnetite,
about 100 nm, are single domain magnetic particles, Where Which contributes to the overall magnetic moment of the
each particle is a magnetic dipole. cobalt-containing magnetic particle 642. In general, the
preferred siZe of the bioprobe 690 may be about 0.1 nm to
[0060] The mechanisms responsible for energy loss exhib about 250 nm, depending upon the disease indication and
ited by single domain particles exposed to an alternating bioprobe composition.
magnetic ?eld are still not Well understood, hoWever a
currently accepted description exists, Which is included [0064] While determining the siZe of the magnetic particle
herein for clarity. When a single domain particle is exposed 642, its material composition may be determined, based on
to an AMF, the Whole magnetic dipole rotates in response to the particular targeted tissue. Because the self-limiting tem
the ?eld With a concomitant energy loss liberated as heat. perature of a magnetic material, or the Curie temperature, is
This mechanism is often referred to as the Neél mechanism. directly related to the material composition, as is the total
The external magnetic forces required for this intrinsic heat delivered, magnetic particle compositions may be tuned
change in magnetiZation depend upon the anisotropy energy to different tissue types. This may be required because each
of the magnetic domain, siZe, and shape of the single domain tissue type, given its composition and location Within the
particle. Furthermore, it is currently accepted that there is a body, possesses unique heating and cooling capacities. For
mechanical rotation of the entire single domain particle example, a tumor located Within a region that is poorly
When exposed to an alternating magnetic ?eld. This latter supplied by blood and located Within a relatively insulating
phenomenon, commonly called the BroWnian mechanism, region may require a loWer Curie temperature material than
also contributes to the energy loss of a single domain a tumor that is located near a major blood vessel. Thus, in
particle, and is proportional to the viscosity of the material addition to magnetite, particle compositions may contain
surrounding the particle. Thus, the coating 644 may enhance elements such as cobalt, iron, rare earth metals, etc.
the heating properties of the bioprobe 690, particularly if the [0065] The presence of the coating 644 and the composi
coating is a polymer. tion of the coating material may form an integral part of the
[0061] The heating mechanism responsible for the energy energy loss, and thus the heat produced, by the bioprobes
loss experienced by a single domain particle in an AMF can 690. In addition, the coating 644 surrounding the particles
be clearly distinguished from the hysteresis heating of larger, may serve additional purposes. Its most important role may
or multidomain magnetic particles. Single domain particles be to provide a biocompatible layer separating the magnetic
of a given composition can produce substantially more heat material from the immunologic defenses in a patient,
per unit mass than multi-domain particles that are 1000 thereby controlling the residence time of the particles in the
times larger (multi domain particles). The heating mecha blood or tissue ?uids.
nism exhibited by single domain particles may be optimiZed [0066] For example, this control of residence time alloWs
to produce superior heating properties over larger particles one to choose targeting ligands 640 that are best suited for
for disease treatment. The amount of heat delivered to a cell a particular tissue type. In addition, the coating 644 may
may be tailored by controlling both the particle siZe and serve to protect the patient from potentially toxic elements
coating variation, as Well as characteristics of the magnetic in the magnetic particle 642. A second function of the
?eld, thereby providing a range of possible bioprobe com coating materials may be the prevention of particle aggre
positions designed for tissue-speci?c treatments. gation, as the bioprobes 690 may be suspended in a ?uid. It
may be also be advantageous to coat the bioprobe 690 With
[0062] Many aspects of the magnetic particle 642, such as
a biocompatible coating that is biodegradable. In such an
material composition, siZe, and shape, directly affect heating
properties. Many of these characteristics may be designed application, both the coating 644 and the magnetic particle
simultaneously to tailor the heating properties for a particu 642 may be digested and absorbed by the body.
lar set of conditions found Within a tissue type. For example, [0067] Suitable materials for the coating 644 include syn
?rst considering the magnetic particle 642, the most desir thetic and biological polymers, copolymers and polymer
able siZe range depends upon the particular application and blends, and inorganic materials. Polymer materials may
on the material(s) comprising the magnetic particle 642. include various combinations of polymers of acrylates,
[0063] The siZe of the magnetic particle 642 determines
siloxanes, styrenes, acetates, akylene glycols, alkylenes,
the total siZe of the bioprobe 690. Bioprobes 690 that are to
alkylene oxides, parylenes, lactic acid, and glycolic acid.
Further suitable coating materials include a hydrogel poly
be injected may be spherical and may be required to have a mer, a histidine-containing polymer, and a combination of a
long residence time in the bloodstream, i.e., avoid seques hydrogel polymer and a histidine-containing polymer.
tration by the liver and other non-targeted organs. The
bioprobe 690 may be successful in avoiding sequestration if [0068] Coating materials may include combinations of
its total diameter is less than about 30 nm. If the bioprobe biological materials such as a polysaccharide, a polyami
690 contains a magnetite (Fe3O4) particle 642, then a noacid, a protein, a lipid, a glycerol, and a fatty acid. Other
preferred diameter of the magnetic particle 642 may be biological materials for use as a coating material may be a
betWeen about 8 nm and about 20 nm. In this case, the heparin, heparin sulfate, chondroitin sulfate, chitin, chitosan,
bioprobes 690 may be sufficiently small to evade the liver, cellulose, dextran, alginate, starch, carbohydrate, and gly
and yet the magnetic particle 642 still retains a suf?cient cosaminoglycan. Proteins may include an extracellular
magnetic moment for heating. Magnetite particles larger matrix protein, proteoglycan, glycoprotein, albumin, pep
than about 8 nm generally tend to be ferrimagnetic and thus tide, and gelatin. These materials may also be used in
appropriate for disease treatment. If other elements, such as combination With any suitable synthetic polymer material.
US 2003/0028071 A1 Feb. 6, 2003

[0069] Inorganic coating materials may include any com the coating 644 in a preferable orientation, speci?cally With
bination of a metal, a metal alloy, and a ceramic. Examples the active region of the ligand 640 available for targeting.
of ceramic materials may include a hydroXyapatite, silicon Physical interaction does not require the linking molecule
carbide, carboXylate, sulfonate, phosphate, ferrite, phospho and the ligand 640 be bound directly to the magnetic particle
nate, and oXides of Group IV elements of the Periodic Table 642 or to the coating 644 by non-covalent means such as, for
of Elements. These materials may form a composite coating eXample, absorption, adsorption, or intercalation.
that also contains any biological or synthetic polymer. [0073] FIG. 7 schematically shoWs an eXample of a ligand
Where the magnetic particle 642 is formed from a magnetic that may be used With an embodiment of the present
material that is biocompatible, the surface of the particle invention. The ligand may be an antibody having a fragment
itself operates as the biocompatible coating. crystalliZation (Fc) region 760 and fragment antigen binding
[0070] The coating material may also serve to facilitate (Fab) regions 772. The Fab regions 772 may be the antigen
transport of the bioprobe 690 into a cell, a process knoWn as binding regions of the antibody that include a variable light
transfection. Such coating materials, knoWn as transfection region 764 and a constant light region 766 along With a
agents, include vectors, prions, polyaminoacids, cationic variable heavy region 768 and a constant heavy region 770.
liposomes, amphiphiles, and non-liposomal lipids or any Biological activity of antibodies may be determined to a
combination thereof. A suitable vector may be a plasmid, a large eXtent by the Fc region 760 of the antibody molecule.
virus, a phage, a viron, a viral coat. The bioprobe coating The Fc region may include complement activation constant
may be a composite of any combination of transfection heavy chains 782 and macrophage binding constant heavy
agent With organic and inorganic materials, such that the chains 784. The Fc region 760 and Fab regions 772 may be
particular combination may be tailored for a particular type connected by several disul?de linkages 762. Ligands that do
of a diseased cell and a speci?c location Within a patient’s not include the Fc region 760 may be preferable in order to
body. avoid immunogenic response. EXamples of these ligands
may include antibody fragments such as, fragment antigen
[0071] To ensure that the bioprobe 690 selectively attaches binding fragments (Fabs) 772, disul?de-stabilized variable
to the diseased cells, an appropriate ligand 640 may be region fragments (dsFVs) 774, single chain variable region
combined With the bioprobe 690. The association of a ligand fragments (scFVs) 780, recombinant single chain antibody
or ligands With the bioprobes 690 alloWs for targeting of fragments, and peptides.
cancer markers on cells. The term ligand relates to com
pounds Which may target molecules including, for eXample, [0074] An antigen binding fragment (Fab) 772 may
proteins, peptides, antibodies, antibody fragments, saccha include a single Fab region 772 of an antibody. Asingle Fab
rides, carbohydrates, glycans, cytokines, chemokines, nucle region may include a variable light 764 and a constant light
region 766 bound to a variable heavy 768 and a constant
otides, lectins, lipids, receptors, steroids, neurotransmitters,
Cluster Designation/Differentiation (CD) markers, and heavy region 770 by a disul?de bond 762.
imprinted polymers and the like. The preferred protein [0075] A disul?de-stabilized variable region fragment
ligands include, for eXample, cell surface proteins, mem (dsFV) 774 may include a variable heavy region 768 and a
brane proteins, proteoglycans, glycoproteins, peptides and variable light region 764 of antibody joined by a disul?de
the like. The preferred nucleotide ligands include, for bond. A leader sequence 776, Which may be a peptide, may
eXample, complete nucleotides, complimentary nucleotides, be linked to the variable light 764 and variable heavy regions
and nucleotide fragments. The preferred lipid ligands 7 68.
include, for eXample phospholipids, glycolipids, and the [0076] Asingle chain variable region fragment (scFV) 780
like. The ligand 640 may be covalently bonded to or may include a variable heavy region 768 and variable light
physically interacted With the magnetic particle 642 or the region 764 of antibody joined by a linker peptide 778. A
coating 644. The ligand 640 may be bound covalently or by leader sequence 776 may be linked to the variable heavy
physical interaction directly to an uncoated portion of the region 768.
magnetic particle 642. The ligand 640 may be bound
covalently or by physical interaction directly to an uncoated [0077] A preferred ligand embodiment may include, for
portion of the magnetic particle 642 and partially covered by eXample, polyclonal antibodies, monoclonal antibodies, chi
the coating 644. The ligand 640 may be bound covalently or meric antibodies, humaniZed antibodies, human antibodies,
by physical interaction to a coated portion of the bioprobe recombinant antibodies, bispeci?c antibodies, antibody frag
690. The ligand 640 may be intercalated to the coated ments, scFVs 780, Fabs 772, dsFVs 774, recombinant single
portion of the bioprobe 690. chain antibody fragments, peptides, and the like. Bispeci?c
antibodies are non-natural antibodies that bind tWo different
[0072] Covalent bonding may be achieved With a linker epitopes that are typically chosen on tWo different antigens.
molecule. The term “linker molecule,” as used herein, refers Abispeci?c antibody is typically comprised of tWo different
to an agent that targets particular functional groups on the fragment antigen binding regions (Fabs) 772. A bispeci?c
ligand 640 and on the magnetic particle 642 or the coating antibody may be formed by cleaving an antibody into tWo
644, and thus forms a covalent link betWeen any tWo of halves by cleaving the disul?de bonds 762 in the Fc region
these. Examples of functional groups used in linking reac 782 only. TWo antibody halves With different Fab regions
tions include amines, sulfhydryls, carbohydrates, carboXyls, 772 are then combined to form a bispeci?c antibody With the
hydroXyls and the like. The linking agent may be a homo typical “Y” structure. One or more ligands can be present in
bifunctional or heterobifunctional crosslinking reagent, for the bioprobe formulation. Antibodies of virtually any origin
eXample, carbodiimides, sulfo-NHS esters linkers and the may be used according to this embodiment, provided they
like. The linking agent may also be an aldehyde crosslinking bind the cancer marker target, although human, chimeric,
reagent such as glutaraldehyde. The linking agent may be and humaniZed antibodies may aid in avoiding the patient’s
chosen to link the ligand 640 to the magnetic particle 642 or immunogenic response.
US 2003/0028071 A1 Feb. 6, 2003

[0078] In another embodiment, the ligand 640 may be mucin, CD277), is a high molecular Weight, transmembrane
designed to target a speci?c cancer cell marker or markers. mucin glycoprotein expressed by most glandular and epi
The particular cancer cell marker and ligand(s) 640 may be thelial cell lineages. In addition, MUC-l has a large extra
speci?c to, but not limited to, the type and location of the cellular domain, portions of Which may be shed into the
cancer such as, for example, tumors, metastatic cancer, bloodstream. MUC-l may have a protective role, as its
minimal residual disease and the like. The ligand(s) 640 may extracellular domain forms elongated rigid structures
have an af?nity for the cancer marker or markers of interest. extending above other molecules on the cell. MUC-l also
The cancer marker or markers may be selected such that they plays a role in cell-cell and cell-substrate adhesion. MUC-l
represent a viable target on the cancer cells of interest. The
is highly expressed in many human adenocarcinomas,
preferred cancer marker may be expressed on the surface of including 80% of breast cancers, and is associated With poor
the cancer cells and is preferably present in very loW
prognosis. Mucin (MUC-l and MUC-2) expression is asso
amounts or not at all in normal cells. The preferred cancer
ciated With tumor invasiveness. MUC-l and MUC-2 expres
marker may not be readily shed from the surface, or if shed, sion is associated With invasive ductive carcinoma of the
the ligand on the bioprobe may recognize a particular breast. MUC-l is also present at high levels on many
epitope of the marker that remains on the cell surface. mylomas. Different tissues/cells produce differing glyco
[0079] FIG. 8 discloses an embodiment of the present forms of MUC-l. Glycosylation of MUC-l in malignant
invention Wherein the bioprobes 890 include a magnetic cells is often altered compared to normal tissue. MUC-l is
particle 842 and a coating material 844 and are bound to the considered a truly tumor speci?c antigen, although it is also
cancer cell surface (Wall) 846 by the ligand 840. The cell found on normal cells, its aberrant glycosylation on tumors
Wall 846 may express several types of chemical species 848 creates neW epitopes for targeting. The extracellular domain
and 850 that may serve as markers. The speci?city of the of MUC-l may be shed into the blood stream The ligand
bioprobes 890 may be represented by attachment to the may target the unshed remainder of the MUC-l expressed on
targeted cell marker 850 over the many other markers or the cell surface.
molecules 848 on the cell surface 846. One or more bio
probes may be attached to the cell surface 846 via the ligand [0082] Overexpression of groWth factor receptors such as
840. The ligands 840 may be adapted and the bioprobes 890 the EGFR family is indicated in tumors and has been
may be designed such that the bioprobes 890 remain on the associated With increased cell resistance to the cytotoxic
cell surface 846 or they may be internaliZed into the cell. effects of macrophages and cytotoxic factors, such as TNF
Once bound to the cell surface 846, the magnetic particle (tumor necrosis factor), Which can lead to tumor groWth. The
842 of the bioprobe 890 heats in response to the AMP. The protein encoded by the Her-l/neu gene is a 170,000 Dalton
heat may emanate through the coating material 844 or protein, referred to as Her-1. The protein encoded by the
through interstitial regions via convection, conduction, Her-2/neu gene is a 185,000 Dalton protein referred to as
radiation, or any combination of these heat transfer mecha Her-2. Both proteins have an intracellular domain, a trans
nisms to the cell surface 846. The heated cell surface 846 membrane domain that includes tWo cysteine-rich repeat
becomes damaged, preferably in a manner that causes clusters, and an intracellular kinase domain. The extracel
irreparable damage to the cell. When a bioprobe(s) 890 lular domain of Her-2 may be shed into the bloodstream.
becomes internaliZed Within the cell, the bioprobe heats the Thus, the ligand may target the unshed remainder of the
Her-2 expressed on the surface of the cell.
cell internally via convection, conduction, radiation, or any
combination of these heat transfer mechanisms. When a [0083] Amethod of administering the bioprobes 890 to the
sufficient amount of heat is transferred by the bioprobes 890 desired area for treatment and the dosage may depend upon,
to the cell, the cell dies by necrosis, apoptosis or another but is not limited to, the type and location of the cancer. The
mechanism. siZe range of the bioprobes 890 alloWs for micro?ltration for
[0080] The choice of a cancer marker (antigen) 850 may steriliZation. An administration method may be, for
be important to therapy utiliZing bioprobes. For breast example, Wash, lavage, as a rinse With sponge, or other
cancer, a speci?c marker or markers may be chosen from surgical cloth after surgery, or intravascular injection, intra
cell surface markers such as, for example, members of the venous injection, intraperitoneal injection, subcutaneous
MUC-type mucin family, an epithelial groWth factor injection, and intramuscular injection. The bioprobes 890
(EGFR) receptor, a carcinoembryonic antigen (CEA), a may be formulated in an injectable format (suspension,
human carcinoma antigen, a vascular endothelial groWth emulsion) in a medium such as, for example, Water, saline,
factor (VEGF) antigen, a melanoma antigen (MAGE) gene, Ringer’s solution, dextrose, albumin solution, and oils.
family antigen, a T/Tn antigen, a hormone receptor, groWth Delivery of the bioprobes 890 to the target site may be
factor receptors, a cluster designation/differentiation (CD) assisted by an applied static magnetic ?eld due to the
antigen, a tumor suppressor gene, a cell cycle regulator, an magnetic nature of the bioprobes. Assisted delivery may
oncogene, an oncogene receptor, a proliferation marker, an depend on the location of the targeted cancer. The bioprobes
adhesion molecule, a proteinase involved in degradation of may also be delivered to the patient using other methods. For
extracellular matrix, a malignant transformation related fac example, the bioprobes may be administered to the patient
tor, an apoptosis related factor, a human carcinoma antigen, orally, or may be administered rectally.
glycoprotein antigens and DF3, 4P2 and MGFM antigens.
EXAMPLES
[0081] In another embodiment of the invention, a bioprobe
may include ligand(s) targeting the MUC-l receptor of the [0084] Having generally described the invention, a more
mucin family. In yet another embodiment, a bioprobe has complete understanding thereof may be obtained by refer
ligand(s) targeting at least one of the EGFR family such as ence to the folloWing examples that are provided for pur
Her-1, Her-2, Her-3 and Her-4. MUC-l (Human epithelial poses of illustration only and do not limit the invention.
US 2003/0028071 A1 Feb. 6, 2003

Example 1 ylmethacrylate (5.38><10_4 moles), N,N‘-methylene bisacry


lamide (§.12><10_5 moles), and 2,2‘-aZobisbutyronitrile
Identify Ferromagnetic Particles With Proper Curie (7.62><10 moles) are added to a mixture of Water/toluene/
Temperatures bis-2-ethylhexyl sulfosuccinate (0.38/0.47/5.1><10_2 moles)
With magnetic particles. The polymeriZation may be carried
[0085] For magnetic materials, the saturation magnetiZa out at 55° C. for several hours under nitrogen. Coated
tion (4J'EMS) and the Curie temperatures (To) depend on the bioprobe particles may then be recovered by precipitation in
material composition. The Curie temperature of the material
an excess of acetone/ethanol folloWed by several Washings.
may lie in the range 40° C.-150° C., although it may also lie
The coated bioprobe particles may be kept in vacuum
outside this range. For example, manganese alloys of the
overnight to remove residual monomers.
general formula RMnZX, Where R is a rare earth, such as La,
Ce, Pr, or Nb, and X is either Ge or Si, exhibit Tc’s of 30-60° Example 3
C. and may be used in bioprobes. Other oxides of ferrite,
garnets, and spinels may be used as bioprobes. For example, Attachment of the Antibodies to the Particles
substituting (doping) Gd and Al for Fe of Y3Fe5O12 garnets
provides control of both Tc and 4J'IZMS. These types of [0090] The attachment of the antibodies to the coated
ferromagnetic media are commercially available from, for magnetic particles may be accomplished by use of a small
example, TRAK Ceramics, HagerstoWn, Md.; Ceramic “linker” molecule With differentiated terminals that permit
Magnetics, Fair?eld, N.J.; and TMC Magnetics, Pine Brook, covalent antibody attachment to the biocompatible polymer
N.J. Sub-micron siZe particles may be produced through ball coating. Attachment occurs in the Fc region using the
milling, solution-precipitation, or sol-gel processing of these electrophilic C-terminal residue of the antibody to react With
materials. the nucleophilic portion of the linker. The other terminus of
the linker may be activated by UV photolysis. The photo
[0086] The heating pro?le (temperature versus time) of the active end of the linker may react With the polymer coating.
magnetic material may be determined under a variety of Conjugation of the antibodies to the nanoparticles using this
model conditions, for example, as suspensions in Water, technique may proceed in the folloWing manner. The anti
Within cell culture media, agar gel, etc. body may be covalently conjugated to 4-[p-aZidosalicyla
[0087] In general, a fundamental frequency of several mido]butylamine (ASBA) using 1-ethyl-3-(3-dimethylami
MHZ is effective for inductive heating. At such frequencies, nopropyl)-carbodiimide, EDC. The EDC may activate the
hoWever, tissue heating may be problematic. Another fre C-terminal carboxyl function, making it susceptible to attack
quency range, Which results in less tissue heating, is 100 kHZ by the primary amino group of the ASBA. The antibody,
to 500 kHZ. covalently tethered to the linker, may be subjected to pho
tolysis in the presence of the polymer-coated nanoparticles.
[0088] Tissue heating places limitations on the practical On exposure to UV, the aZido function on the linked ASBA
amplitude of the magnetic ?eld. A preferred range may be may generate a nitrene in-situ that covalently inserts into the
from 100 to 200 Oersteds (Oe), using a constant Waveform polymer coat yielding a nanoparticle that may be covalently
generator. Dif?culties may arise in achieving desired particle bound to the antibody through the linker.
heating With many materials When the ?eld falls in this
range. The ?eld conditions may also be inadequate because [0091] If the selected magnetic nanoparticles are not toxic
the number of bioprobes attaching to the targeted cells may and do not require polymer coating, the antibodies may be
often be unknoWn and/or limited by the speci?cs of cellular attached to the nanoparticles directly. The immunoglobulin
biology. The limitation may be overcome by using a pulsed G’s may be cleaved at the disul?de linkages connecting the
generator and setting the ?eld conditions, ?eld amplitude tWo portions of the antibody and puri?ed by chromatogra
and pulse characteristics, to levels that heats the bioprobes phy. The fragments, containing residual sulfhydryl groups,
sufficiently to kill targeted cells Without excessive peripheral may be conjugated directly to the metal clusters forming
tissue heating. Thus, under pulsed conditions, peak magnetic coordinate covalent linkages.
?elds of up to a feW thousand Oersteds (Oe) may be used.
Example 4
Example 2
Ef?cacy of Bioprobes: In vitro Trials With MCF-7
Synthesis of Bio-Compatible Coating Breast Cancer Cells

[0089] The bioprobe particles selected during EXAMPLE [0092] The bioprobes included 50 nm, Fe3O4 particles
surrounded by a dextran shell, to Which the monoclonal
1 may be coated With a biocompatible polymer according to
antibody for Her-2 Was covalently linked. The
a folloWing embodiment of the procedure. Poly (methacrylic
particles Were suspended in cellular groWth media including
acid-co-hydroxyethylmethacrylate) as a biocompatible coat
ing material for bioprobes may be synthesiZed from meth Modi?ed Eagle’s Medium (MEM) containing 10% fetal calf
acrylic acid and hydroxyethyl methacrylate using free-radi serum, insulin (10 pig/ml), fungiZone, glutamine, penicillin,
streptomycin, sodium pyruvate and non-essential amino
cal polymeriZation in the presence of the magnetic particles.
acids, Which Was then added to cultures of MCF-7 cells. The
To avoid aggregation of the magnetic nanometer siZed
MCF-7 cells represent an estrogen receptor-positive human
particles during polymeriZation, the polymeriZation may be breast carcinoma, and Were groWn on tissue culture inserts
carried out in a microemulsion environment consisting of
With a 10 mm diameter possessing porous frits With pore
Water/toluene/sodium bis-2-ethylhexyl sulfosuccinate. The siZes of 0.02 or 0.2 pm.
sodium bis-2-ethylhexyl sulfosuccinate acts as an ionic
surfactant to make a stabiliZing layer around the magnetic [0093] The alternating magnetic ?eld source Was an indus
particles. Methacrylic acid (1.45><10_2 moles), hydroxyeth trial 35 kW variable duty radio-frequency generator With a
US 2003/0028071 A1 Feb. 6, 2003

frequency of 740 kHZ and time-averaged ?eld amplitude of presence of the bioprobes, is toxic to the cells. Higher than
500 Oe (peak ?eld amplitude, 1300 Oe). The generator normal cell death occurs only When the AME is applied after
produced pulses With a length of 4.174 ms, at a pulse the bioprobes have attached to a cell.
repetition rate of 121 HZ, to give a duty cycle of 50.6%. The
generator provided poWer to a 14 mm (inner diameter), TABLE 2
S-turn copper solenoid coil into Which sample containers
Were inserted. The average temperature of the media in all IN VITRO RESULTS WITH MCF-7 (‘PI I S

samples monitored in situ using silicon dioxide temperature SAMPLE TREATMENT % DEAD CELLS (N = 7)
probes resistant to electromagnetic (EM) ?elds (obtained
from FISO Technologies Inc., Ste-Foy, Quebec, Canada). C1 NO BIOPROBES, NO AMF 4 I 1
C2 BIOPROBES, NO AMF 5 I 1
[0094] A series of Water bath tests Was conducted to C3 NO BIOPROBES; AMF 4 I 1
T1 BIOPROBES WITH AMF 91 I 5
determine the hyperthermia tolerance of the cell cultures.
The purpose of these tests Was to obtain “positive control”
data on the effects of hyperthermia on the cancer cell
cultures and to aid in the interpretation of experiments using Example 5
the bioprobe system. Exposing cellular culture media to an
alternating magnetic ?eld, Without the presence of the bio Selectivity of Bioprobes: In vitro Trials With
probes, may cause some tissue heating. Such magnetic SK-CO-1 Colon Cancer Cells
induction heating is determined by the frequency and ?eld
[0096] For the in vitro studies, the MCF-7 human breast
strength of the magnetic ?eld. Thus, to avoid overheating the cancer cells Were chosen because their Her-2 expression Was
media With the applied magnetic ?eld, Water bath tests Were
suf?cient for detection, but Was not expressed in extraordi
conducted to provide threshold limits of magnetic induction narily high amounts as is the case With the more aggressive
heating of the media containing cells not receiving the MDA-MB-231 (human breast carcinoma cells knoWn to
treatment. Table 1 lists results of cell death fraction as a
signi?cantly over express Her-2) cell line. As a control,
function of exposed temperature and exposure time. The cultures of SK-CO-1 cells (human colon adenocarcinoma
MCF-7 cell line is shoWn to be unaffected by heat exposure
cells knoWn to be Her-2 negative) Were chosen to provide a
to 42° C. for 30 minutes. Thus, for all in vitro experiments, reasonable model to challenge the effectiveness and selec
the magnetic ?eld strengths Were ?xed at levels Where the
tivity of the bioprobe system.
average temperature of the cellular groWth media (contain
ing no cells or magnetic ?uid) remained at or beloW 42° C. [0097] To investigate the selectivity of the bioprobes,
SK-CO-1 human colon cancer cells knoWn to be Her-2
TABLE 1 negative Were treated in the same manner. For this group,
rates of apoptosis Were 13% for cells treated With an
Result of Water bath tests With MCF-7 cells alternating magnetic ?eld and bioprobes, 18% for
Temperature (O C.) Time of exposure (min) % dead cells
cells receiving no AMP and no bioprobes (represents normal
death rate), 10% for cells exposed only to an AMP, and 8%
37* N/A 4 for cells exposed to only bioprobes. These results are
42 30 4
43 15 5
summariZed in Table 3. Thus, no signi?cant differences Were
46 5 92 seen in the death rates of SK-CO-1 cells When used With any
or all components of the invention, demonstrating selective
*Represents a control cell culture sample not placed into Water bath. tumoricidal activity for the MCF-7 breast cells.

[0095] Cells intended for treatment Were combined With TABLE 3


bioprobes containing the Her-2 antibody and incubated for
IN VITRO RESULTS WITH SK-CO-1 CELLS
8 minutes at 20° C., folloWed by three rinses With groWth
media, to remove unattached bioprobes. The cell cultures SAMPLE TREATMENT % DEAD CELLS
Were analyZed before and 6 hours after the 20 minute
treatments With the alternating magnetic ?eld The SK-CO-1 NO BIOPROBES, NO AMP 18
SK-CO-1 BIOPROBES, NO AMF 8
fraction of dead cells for the targeted sample (T1) and SK-CO-1 NO BIOPROBES; AMP 13
control samples (C1-C3) are presented in Table 2. In the T1 (MCF-7) BIOPROBES WITH AMP 91 r 5 (N = 7)
targeted sample, 91%:5% (n=7) of the MCF-7 cells Were
killed. Of the cells killed, about 70% cells Were be lysed by
the treatment, as measured by spectrophotometric analysis Example 6
of cytoplasmic lactate dehydrogenase (LDH), an enZyme
produced by living cells. The remaining approximately 20% Bioprobe Targeting MUC-1 Receptor in Breast
underWent apoptosis, as measured using a commercial ?uo
Cancer
rescent apoptosis-staining assay. The kill rates for the tar
geted cells are signi?cantly higher than baseline death and [0098] The target on the breast cancer cells may be
apoptotic rates of 4%:1% in all controls (Table 2). Control MUC-1 (Human epithelial mucin, CD277) marker. MUC-1
groups include: 1) cells receiving no exposure to either the is highly expressed in many human adenocarcinomas
AME or bioprobes (Sample C1); 2) cells exposed to the including 80% of breast cancers and is associated With poor
bioprobes alone (Sample C2); and 3) cells exposed to the prognosis. The ligand on the bioprobe may be an antibody
AME alone (Sample C3). Similar baseline deaths of all Which targets the MUC-1 receptor. Commercial monoclonal
controls con?rm that neither the AME alone, nor only the mouse anti-human MUC-1 antibodies may be used in pre
US 2003/0028071 A1 Feb. 6, 2003

liminary in vitro studies, from such sources as Chemicon none of the probes 354 sense a temperature greater than the
International, Temecula, Calif; and Zymed Laboratories, preset limit, the RF generator 318 may be permitted to
South San Francisco, Calif. The MUC-l bioprobes may be operate at the original settings. If one or more of the probes
formed from 50 nm dextran coated Fe3O4 particles. The 354 senses a temperature over the preset threshold, the
mouse anti-human MUC-l is a Whole antibody and the Fc controller 356 may send a command to reduce at least one
portion may be linked to the magnetic particle or the dextran of the duty cycle, the PRF, the magnitude of the magnetic
coating through several different techniques. A linking ?eld. This process continues until the treatment is com
agent, such as BS3, available from Pierce, Rockford, Ill., pleted.
may be used With silaniZation or the antibody may be
[0102] While the above description of the invention has
biotinylated and attached to a streptavidin-containing, 50 nm
been presented in terms of a human patient, it Will be
dextran coated particle.
appreciated that the invention may also be applicable to
[0099] In vitro studies demonstrate dose response and treating cancers in other mammals.
proof of killing of cancer cells above the level of the
controls. The targeted bioprobes may be further re?ned by [0103] As noted above, the present invention is applicable
to a magnetic material composition, a system and method of
producing Fab and scFV fragments of a monoclonal mouse
thermotherapy for the treatment of cancers. The present
anti-human MUC-l antibody, fragments from an antibody
invention should not be considered limited to the particular
library, humaniZation of the antibody, or production of
examples described above, but rather should be understood
peptides. Antibodies, fragments, and peptides may be engi to cover all aspects of the invention as fairly set out in the
neered With altered properties such as antigen binding
af?nity, molecular architecture, and speci?c receptor epitope attached claims. Various modi?cations, equivalent pro
cesses, as Well as numerous structures to Which the present
targeting to enhance the tumor targeting and potency. The
invention may be applicable Will be readily apparent to those
use of fragments, humaniZed antibody, or peptides creates a
of skill in the art to Which the present invention is directed
ligand that avoids immunogenic response in humans.
upon revieW of the present speci?cation. The claims are
[0100] The MUC-l receptor is considered-a truly tumor intended to cover such modi?cations and devices.
speci?c antigen because although it is on normal cells, its
aberrant glycosylation on tumors creates neW epitopes for What is claimed is:
targeting. The extracellular domain of MUC-l is shed into 1. A magnetic material composition, comprising:
the blood stream Which makes the selection of the targeting
ligand a very important component in this example. The a) a particle having magnetic properties and forming a
MUC-l targeting ligand may be selected to avoid the shed single magnetic domain;
portion of the extracellular domain of MUC-l in the blood
b) a biocompatible coating material for the particle; and
stream. This may result in reducing delivery problems and
dilution of the treatment by bioprobes binding to the shed c) a ligand selective to at least one cancer marker on a cell
receptor. The MUC-l targeting ligand recogniZes an epitope in cancer tissue, the ligand being i) bound to an
of MUC-l Which remains on the cancer cell after a portion uncoated portion of the particle, ii) bound to a coated
of the extracellular domain is shed. This choice of ligand portion of the particle, iii) bound to the particle and
alloWs the bioprobe to speci?cally and selectively target partially covered by the coating or iv) intercalated into
breast cancer cells. the coating.
2. A magnetic particle composition of claim 1, Wherein
Example 7 the biocompatible coating material is biodegradable.
3. A magnetic particle composition of claim 1, Wherein
Temperature Monitoring During Treatment the particle has a siZe of no more than 250 nm in at least one
dimension.
[0101] In a treatment cycle, the bioprobes 210 may be
exposed to and become attached to metastatic cells in the 4. A magnetic material composition of claim 1, Wherein
treatment area. Pole pieces 204 may be chosen to provide the particle, the coating and the ligand are suspended in a
treatment in a selected treatment area, for example, approxi
biologically compatible ?uid.
mately 6 inches in diameter. The patient 105 is typically 5. A magnetic material composition of claim 1, Wherein
placed on the treatment bed 206, and the bed controller 108 the magnetic particle is ferromagnetic, antiferromagnetic,
is typically adjusted to place the targeted tissue at the region ferrimagnetic, antiferrimagnetic or superparamagnetic.
of maximum ?eld strength betWeen the poles 204. The pulse 6. A magnetic material composition of claim 1, Wherein
Width, duty cycle, and peak amplitude of the AMF and the the magnetic particle has a Curie temperature in the range of
treatment time may be adjusted. Several temperature probes, about 40° C. to about 150° C.
for example, four temperature probes, may be inserted into 7. A magnetic material composition of claim 1, Wherein
the treatment volume. One probe may be located centrally the magnetic particle is formed of a biocompatible material,
and the other three may be inserted into tissues, Within the the surface of the magnetic particle forming the biocompat
treatment area that are heated by the AMF. As the tissue ible coating.
heats under the applied AMF, the temperature probes 354 8. A magnetic material composition of claim 1, Wherein
send data to the probe monitor 352. The monitor sends the the biocompatible coating material is one of an organic
temperature data via the feedback loop 324 to the RF material, an inorganic material, and a combination of an
generator controller 356. Monitoring may be continuous, for inorganic material and an organic material.
example at a data rate of 10 samples per second and the duty 9. A magnetic material composition of claim 8, Wherein
of the RF generator 318 may be adjusted once per second the organic material is at least one of a synthetic material and
based on individual, 10 sample averages from each probe. If a biological material.
US 2003/0028071 A1 Feb. 6, 2003

10. A magnetic material composition of claim 9, wherein ine, xi) a chemokine, xii) a nucleotide, xiii) a lipid, xiv) a
the synthetic material is a polymer, a copolymer, or a steroid, xv) a neurotransmitter, xvi) a lectin, xvii) an
combination thereof. imprinted polymer, xviii) an oncogene, xix) an oncogene
11. A magnetic material composition of claim 9, Wherein receptor and xx) a combination of any of i)-xix).
the synthetic material includes at least one of i) a polymer, 26. Amagnetic material composition of claim 25, Wherein
ii) a copolymer, and iii) a polymer blend formed from a the protein includes one of a) a membrane protein, b) a
polymer based on at least one of acrylates, siloxanes, proteoglycan, c) a cell surface protein, d) a glycoprotein, and
styrenes, acetates, alkylene glycols, alkylenes, alkylene e) a combination of any of a)-e).
oxides, parylene, lactic acid, and glycolic acid. 27. Amagnetic material composition of claim 25, Wherein
12. A magnetic material composition of claim 9, Wherein the antibody includes one of a) a polyclonal antibody, b) a
the synthetic material includes one of i) a hydrogel polymer, monoclonal antibody, c) a chimeric antibody, d) a human
ii) a histidine-containing polymer, iii) a surfactant and iv) a iZed antibody, e) a human antibody, f) a recombinant anti
combination of at least one of i)-iii). body, g) a bispeci?c antibody, h) an antibody fragment, i) a
13. A magnetic material composition of claim 9, Wherein recombinant single chain antibody fragment and a com
the biological material includes one of i) a polysaccharide, bination of any of a-i)
ii) a polyaminoacid, iii) a protein, iv) a lipid, v) a glycerol, 28. Amagnetic material composition of claim 25, Wherein
vi) a fatty acid, and vii) a combination including at least one the nucleotide includes a) a complete nucleotide, b) a
of i)-vi). nucleotide fragment, c) a complementary nucleotide, and d)
14. Amagnetic material composition of claim 13, Wherein a combination of a)-c).
the polysaccharide includes one of a) a heparin, b) heparin 29. Amagnetic material composition of claim 25, Wherein
sulfate, c) chondroitin sulfate, d) chitin, e) chitosan, f) the lipid includes a) a phospholipid, b) a glycolipid, or c) a
cellulose, g) dextran, h) alginate, i) starch, saccharide, k) combination of a) and b).
carbohydrate, l) glycosaminoglycan and m) a combination 30. Amagnetic material composition of claim 24, Wherein
of any of a)-l). the marker is speci?c to breast cancer.
15. Amagnetic material composition of claim 13, Wherein 31. Amagnetic material composition of claim 29, Wherein
the protein includes one of a) an extracellular matrix protein,
the marker is speci?c to a metastatic cancer related to breast
b) proteoglycan, c) glycoprotein, d) albumin, e) peptide, f) cancer.
gelatin, and g) a combination of any of a)-f).
16. A magnetic material composition of claim 8, Wherein 32. Amagnetic material composition of claim 29, Wherein
the marker is speci?c to a primary cancer of the breast.
the inorganic material includes one of i) a metal, ii) a metal
alloy, iii) a ceramic, iv) an oxide of a Group IV element, and 33. Amagnetic material composition of claim 29, Wherein
v) a combination of any of i)-iv). the breast cancer-speci?c marker includes one of a) a
17. Amagnetic material composition of claim 16, Wherein member of the MUC-type mucin family, b) a member of the
the ceramic includes one of a) hydroxyapatite, b) silicon epidermal groWth factor receptor (EGFR) family, c) a car
carbide, c) carboxylate, d) sulfonate, e) phosphate, f) ferrite, cinoembryonic antigen (CEA), d) a MAGE (melanoma
g) phosphonate, and h) a combination of any of a)-g). antigen) gene family antigen, e) a T/Tn antigen, f) a hor
18. A magnetic material composition of claim 9, Wherein mone receptor, g) a Cluster Designation/Differentiation
the biological material is a transfection agent to enhance (CD) antigen, h) a tumor suppressor gene, i) a cell cycle
uptake by cancer cells. regulator, an oncogene, k) an oncogene receptor, 1) a
19. Amagnetic material composition of claim 18, Wherein proliferation marker, m) an adhesion molecule, n) a protein
ase involved in degradation of extracellular matrix, 0) a
the transfection agent includes one of i) a vector, ii) a prion,
iii) a polyaminoacid, iv) a cationic liposome, v) an malignant transformation related factor, p) an apoptosis
amphiphile, vi) a non-liposomal lipid, and vii) a combina related factor, q) a human carcinoma antigen, r) a member of
tion of any of i)-vi).
the vascular endothelial groWth factor (VEGF) receptor
20. Amagnetic material composition of claim 19, Wherein family, s) glycoprotein antigens, t) DF3 antigen, u) 4F2
antigen, v) MFGM antigen and W) a combination of any of
the vector includes one of a) a plasmid, b) a virus, c) a phage,
d) a viron, e) a viral coat, and f) a combination of any of
a) through v).
34. Amagnetic material composition of claim 32, Wherein
a)-e). the MUC-type mucin family marker is MUC-l.
21. Amagnetic material composition of claim 19, Wherein
the polyaminoacid is a poly L-lysine. 35. Amagnetic material composition of claim 32, Wherein
22. A magnetic material composition of claim 9, Wherein the EGFR includes at least one of Her-1, Her-2, Her-3,
the synthetic material is a transfection agent. Her-4, and any combination thereof.
23. Amagnetic material composition of claim 22, Wherein 36. A magnetic material composition of claim 1, further
the transfection agent includes one of i) a non-lipid cationic comprising a linking agent linking betWeen the ligand and
polymer, ii) a dendrimer, iii) polyethyleneimine, and iv) a the magnetic particle or betWeen the ligand and the biocom
combination of any of i)-iii). patible coating material.
24. A magnetic material composition of claim 1, Wherein 37. Amagnetic material composition of claim 35, Wherein
the ligand is one of a molecule and a combination of the linking agent links to at least one of i) an amine group,
molecules selective to at least one speci?c cancer marker. ii) a sulfhydryl group, iii) a carbohydrate group, iv) a
25. Amagnetic material composition of claim 24, Wherein carboxyl group, v) a hydroxyl group, and vi) a combination
the molecule includes one of i) a saccharide, ii) a carbohy of any of i)-v).
drate, iii) a glycan, iv) a protein, v) a peptide, vi) an 38. Amagnetic material composition of claim 35, Wherein
antibody, vii) an antibody fragment, viii) a receptor, ix) a the linking agent is comprised of at least one of i) homobi
Cluster Designation/Differentiation (CD) marker, X) a cytok functional crosslinkers, ii) heterobifunctional crosslinkers,

Vous aimerez peut-être aussi